EP1064382B1 - Polypeptides homologues aux vegf et bmp1 - Google Patents

Polypeptides homologues aux vegf et bmp1 Download PDF

Info

Publication number
EP1064382B1
EP1064382B1 EP99912362A EP99912362A EP1064382B1 EP 1064382 B1 EP1064382 B1 EP 1064382B1 EP 99912362 A EP99912362 A EP 99912362A EP 99912362 A EP99912362 A EP 99912362A EP 1064382 B1 EP1064382 B1 EP 1064382B1
Authority
EP
European Patent Office
Prior art keywords
vegf
polypeptide
cells
cell
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP99912362A
Other languages
German (de)
English (en)
Other versions
EP1064382A2 (fr
Inventor
Napoleone Ferrara
Sophia S. Kuo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/040,220 external-priority patent/US6391311B1/en
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to DK08012659.2T priority Critical patent/DK2016951T3/da
Priority to EP08012659A priority patent/EP2016951B1/fr
Publication of EP1064382A2 publication Critical patent/EP1064382A2/fr
Application granted granted Critical
Publication of EP1064382B1 publication Critical patent/EP1064382B1/fr
Priority to CY20081101286T priority patent/CY1108500T1/el
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/026Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus

Definitions

  • the present invention is directed to polypeptides related to vascular endothelial cell growth factor (hereinafter sometimes referred to as VEGF) and bone morphogenetic protein 1 (hereinafter sometimes referred to as BMP1), termed herein as VEGF-E polypeptides, nucleic acids encoding therefor, methods for preparing VEGF-E, and methods, compositions, and assays utilizing VEGF-E.
  • VEGF vascular endothelial cell growth factor
  • BMP1 bone morphogenetic protein 1
  • FGF basic and acidic fibroblast growth factors
  • PD-ECGF platelet-derived endothelial cell growth factor
  • VEGF vascular endothelial growth factor
  • VEGF heparin-binding endothelial cell-growth factor
  • VEGF 121 is a soluble mitogen that does not bind heparin; the longer forms of VEGF bind heparin with progressively higher affinity.
  • the heparin-binding forms of VEGF can be cleaved in the carboxy terminus by plasmin to release (a) diffusible form(s) of VEGF.
  • the amino acid sequence of the carboxy-terminal peptide identified after plasmin cleavage is Arg 110 -Ala 111 .
  • VEGF Amino terminal "core” protein, VEGF (1-110), isolated as a homodimer, binds neutralizing monoclonal antibodies (4.6.1 and 2E3) and soluble forms of FMS-like tyrosine kinase (FLT-1), kinase domain region (KDR) and fetal liver kinase (FLK) receptors with similar affinity compared to the intact VEGF 165 homodimer.
  • FLT-1 FMS-like tyrosine kinase
  • KDR kinase domain region
  • FLK fetal liver kinase
  • VEGF is useful for treating conditions in which a selected action on the vascular endothelial cells, in the absence of excessive tissue growth, is important, for example, diabetic ulcers and vascular injuries resulting from trauma such as subcutaneous wounds.
  • VEGF restores cells that are damaged, a process referred to as vasculogenesis, and stimulates the formulation of new vessels, a process referred to as angiogenesis.
  • angiogenesis which involves the formation of new blood vessels from preexisting endothelium, is implicated in the pathogenesis of a variety of disorders. These include solid tumors and metastasis, atherosclerosis, retrolental fibroplasia, hemangiomas, chronic inflammation, intraocular neovascular syndromes such as proliferative retinopathies, e.g ., diabetic retinopathy, age-related macular degeneration (AMD), neovascular glaucoma, immune rejection of transplanted corneal tissue and other tissues, rheumatoid arthritis, and psoriasis.
  • proliferative retinopathies e.g ., diabetic retinopathy, age-related macular degeneration (AMD), neovascular glaucoma
  • AMD age-related macular degeneration
  • neovascular glaucoma immune rejection of transplanted corneal tissue and other tissues
  • rheumatoid arthritis rhe
  • angiogenesis appears to be crucial for the transition from hyperplasia to neoplasia, and for providing nourishment to the growing solid tumor.
  • the neovascularization allows the tumor cells to acquire a growth advantage and proliferative autonomy compared to the normal cells. Accordingly, a correlation has been observed between density of microvessels in tumor sections and patient survival in breast cancer as well as in several other tumors.
  • VEGF has been shown to be a key mediator of neovascularization associated with tumors and intraocular disorders.
  • Ferrara et al. Endocr. Rev., supra .
  • the VEGF mRNA is overexpressed by the majority of human tumors examined. Berkman et al., J Clin Invest, 91: 153-159 (1993 ); Brown et al., Human Pathol., 26: 86-91 (1995 ); Brown et al., Cancer Res., 53: 4727-4735 (1993 ); Mattern et al., Brit. J. Cancer, 73: 931-934 (1996 ); Dvorak et al., Am J. Pathol., 146: 1029-1039 (1995 ).
  • this invention provides a process for producing a VEGF-E polypeptide comprising culturing the host cell described above under conditions suitable for expression of the VEGF-E polypeptide and recovering the VEGF-E polypeptide from the cell culture. Further provided is a polypeptide produced by this process.
  • the invention provides a polypeptide comprising the amino acid sequence set forth in SEQ ID NO:2.
  • the invention provides a pharmaceutical product comprising:
  • the invention provides a method for diagnosing a disease or a susceptibility to a disease related to a mutation in a nucleic acid sequence encoding VEGF-E comprising:
  • the invention provides an article of manufacture, comprising:
  • Figures 5A and 5B show, respectively, the effect on HUVEC tube formation of VEGF-E conjugated to poly-his at 1% dilution and of a buffer control (same as in Fig. 4B) at 1 % dilution.
  • the biological activity of native VEGF-E is shared by any analogue or variant thereof that promotes selective growth and/or survival of umbilical vein endothelial cells, induces proliferation of pluripotent fibroblast cells, induces immediate early gene c-fos in human endothelial cell lines, causes myocyte hypertrophy in cardiac cells, inhibits VEGF-stimulated proliferation of adrenal cortical capillary endothelial cells, or which possesses an immune epitope that is immunologically cross-reactive with an antibody raised against at least one epitope of the corresponding native VEGF-E.
  • the human VEGF-E herein is active on rat and mouse cells, indicating conservation across species. Moreover, the VEGF-E herein is expressed at the growth plate region and has been shown to embrace fetal myocytes.
  • angiogenic disorder refers to a disorder that requires treatment with an agent that inhibits angiogenesis, e.g., an angiostatic compound.
  • disorders include, for example, types of cancer such as vascular tumors, e.g ., hemangioma (capillary and cavernous), glomus tumors, telangiectasia, bacillary angiomatosis, hemangioendothelioma, angiosarcoma, haemangiopericytoma, Kaposi's sarcoma, lymphangioma, and lymphangiosarcoma, and tumor angiogenesis.
  • vascular tumors e.g ., hemangioma (capillary and cavernous), glomus tumors, telangiectasia, bacillary angiomatosis, hemangioendothelioma, angiosarcoma, haemangiopericytoma, Kaposi's sarcoma, lymphangio
  • “Hypertrophy”, as used herein, is defined as an increase in mass of an organ or structure independent of natural growth that does not involve tumor formation. Hypertrophy of an organ or tissue is due either to an increase in the mass of the individual cells (true hypertrophy), or to an increase in the number of cells making up the tissue (hyperplasia), or both. Certain organs, such as the heart, lose the ability to divide shortly after birth. Accordingly, "cardiac hypertrophy” is defined as an increase in mass of the heart, which, in adults, is characterized by an increase in myocyte cell size and contractile protein content without concomitant cell division. The character of the stress responsible for inciting the hypertrophy, ( e .
  • the early stage of cardiac hypertrophy is usually characterized morphologically by increases in the size of microfibrils and mitochondria, as well as by enlargement of mitochondria and nuclei. At this stage, while muscle cells are larger than normal, cellular organization is largely preserved. At a more advanced stage of cardiac hypertrophy, there are preferential increases in the size or number of specific organelles, such as mitochondria, and new contractile elements are added in localized areas of the cells, in an irregular manner.
  • cardiac hypertrophy is used to include all stages of the progression of this condition, characterized by various degrees of structural damage of the heart muscle, regardless of the underlying cardiac disorder. Hence, the term also includes physiological conditions instrumental in the development of cardiac hypertrophy, such as elevated blood pressure, aortic stenosis, or myocardial infarction.
  • Myocardial infarction generally results from atherosclerosis of the coronary arteries, often with superimposed coronary thrombosis. It may be divided into two major types: transmural infarcts, in which myocardial necrosis involves the full thickness of the ventricular wall, and subendocardial (nontransmural) infarcts, in which the necrosis involves the subendocardium, the intramural myocardium, or both, without extending all the way through the ventricular wall to the epicardium. Myocardial infarction is known to cause both a change in hemodynamic effects and an alteration in structure in the damaged and healthy zones of the heart.
  • Administration "in combination with” one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration in any order.
  • VEGF-E variants can be prepared.
  • VEGF-E variants can be prepared by introducing appropriate nucleotide changes into the VEGF-E-encoding DNA, or by synthesis of the desired VEGF-E polypeptide.
  • amino acid changes may alter post-translational processes of the VEGF-E polypeptide, such as changing the number or position of glycosylation sites or altering the membrane-anchoring characteristics.
  • Suitable prokaryotes include but are not limited to eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as E. coli .
  • eubacteria such as Gram-negative or Gram-positive organisms
  • Enterobacteriaceae such as E. coli .
  • E. coli strains are publicly available, such as E. coli K12 strain MM294 (ATCC 31,446); E. coli X1776 (ATCC 31,537); E. coli strain W3110 (ATCC 27,325) and K5 772 (ATCC 53,635).
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for VEGF-E-encoding vectors.
  • Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism.
  • the signal sequence may be, e.g ., the yeast invertase leader, alpha factor leader (including Saccharomyces and Kluyveromyces ⁇ -factor leaders, the latter described in U.S. Patent No. 5,010,182 ), or acid phosphatase leader, the C. albicans glucoamylase leader ( EP 362, 179 published 4 April 1990 ), or the signal described in WO 90/13646 published 15 November 1990 .
  • mammalian signal sequences may be used to direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species, as well as viral secretory leaders.
  • Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the VEGF-E polypeptide.
  • S.D. Shine-Dalgarno
  • Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5' or 3' to the VEGF-E coding sequence, but is preferably located at a site 5' from the promoter.
  • Gene amplification and/or expression may be measured in a sample directly, for example, by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA ( Thomas, Proc. Natl. Acad. Sci. USA, 77:5201-5205 (1980 )), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein.
  • antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn may be labeled and the assay may be carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
  • Assays for wound-healing activity include, for example, those described in Winter, Epidermal Wound Healing, Maibach, HI and Rovee, DT, eds. (Year Book Medical Publishers, Inc., Chicago), pp. 71-112 , as modified by the article of Eaglstein and Mertz, J. Invest. Dermatol., 71: 382-384 (1978 ).
  • In vitro assays include induction of spreading of adult rat cardiac myocytes.
  • ventricular myocytes are isolated from a single (male Sprague-Dawley) rat, essentially following a modification of the procedure described in detail by Piper et al., "Adult ventricular rat heart muscle cells” in Cell Culture Techniques in Heart and Vessel Research, H.M. Piper, ed. (Berlin: Springer-Verlag, 1990), pp. 36-60 . This procedure permits the isolation of adult ventricular myocytes and the long-term culture of these cells in the rod-shaped phenotype.
  • Phenylephrine and Prostaglandin F 2 ⁇ have been shown to induce a spreading response in these adult cells.
  • the inhibition of myocyte spreading induced by PGF 2 ⁇ or PGF 2 ⁇ analogs (e.g ., fluprostenol) and phenylephrine by various potential inhibitors of cardiac hypertrophy is then tested.
  • MI myocardial infarction
  • Acute MI is induced in rats by left coronary artery ligation and confirmed by electrocardiographic examination.
  • a sham-operated group of animals is also prepared as control animals.
  • Earlier data have shown that cardiac hypertrophy is present in the group of animals with MI, as evidenced by an 18% increase in heart weight-to-body weight ratio. Lai et al., supra.
  • Treatment of these animals with candidate blockers of cardiac hypertrophy, e.g ., VEGF-E polypeptide provides valuable information about the therapeutic potential of the candidates tested.
  • One further such assay test for induction of cardiac hypertrophy is disclosed in U.S. Pat. No. 5,773,415 , using Sprague-Dawley rats.
  • Such models can be generated by introducing tumor cells into syngeneic mice using standard techniques, e.g ., subcutaneous injection, tail vein injection, spleen implantation, intraperitoneal implantation, implantation under the renal capsule, or orthopin implantation, e . g ., colon cancer cells implanted in colonic tissue. See, e.g ., PCT publication No. WO 97/33551, published September 18, 1997 .
  • Tumor cells can be introduced into animals such as nude mice by a variety of procedures.
  • the subcutaneous (s.c.) space in mice is very suitable for tumor implantation.
  • Tumors can be transplanted s.c. as solid blocks, as needle biopsies by use of a trochar, or as cell suspensions.
  • tumor tissue fragments of suitable size are introduced into the s.c. space.
  • Cell suspensions are freshly prepared from primary tumors or stable tumor cell lines, and injected subcutaneously. Tumor cells can also be injected as subdermal implants. In this location, the inoculum is deposited between the lower part of the dermal connective tissue and the s.c. tissue.
  • Tumors that arise in animals can be removed and cultured in vitro. Cells from the in vitro cultures can then be passaged to animals. Such tumors can serve as targets for further testing or drug screening. Alternatively, the tumors resulting from the passage can be isolated and RNA from pre-passage cells and cells isolated after one or more rounds of passage analyzed for differential expression of genes of interest. Such passaging techniques can be performed with any known tumor or cancer cell lines.
  • the Lewis lung (3LL) carcinoma of mice which is one of the most thoroughly studied experimental tumors, can be used as an investigational tumor model. Efficacy in this tumor model has been correlated with beneficial effects in the treatment of human patients diagnosed with small-cell carcinoma of the lung (SCCL).
  • SCCL small-cell carcinoma of the lung
  • This tumor can be introduced in normal mice upon injection of tumor fragments from an affected mouse or of cells maintained in culture. Zupi et al., Br. J. Cancer, 41: suppl. 4, 30 (1980 ). Evidence indicates that tumors can be started from injection of even a single cell and that a very high proportion of infected tumor cells survive. For further information about this tumor model see Zacharski, Haemostasis, 16: 300-320 (1986 ).
  • One way of evaluating the efficacy of a test compound in an animal model with an implanted tumor is to measure the size of the tumor before and after treatment.
  • the size of implanted tumors has been measured with a slide caliper in two or three dimensions.
  • the measure limited to two dimensions does not accurately reflect the size of the tumor; therefore, it is usually converted into the corresponding volume by using a mathematical formula.
  • the measurement of tumor size is very inaccurate.
  • the therapeutic effects of a drug candidate can be better described as treatment-induced growth delay and specific growth delay.
  • Another important variable in the description of tumor growth is the tumor volume doubling time.
  • Computer programs for the calculation and description of tumor growth are also available, such as the program reported by Rygaard and Spang-Thomsen, Proc. 6th Int.
  • nucleic acids that encode VEGF-E polypeptide or any of its modified forms can also be used to generate either transgenic animals or "knock-out" animals which, in turn, are useful in the development and screening of therapeutically useful reagents.
  • a transgenic animal e.g., a mouse or rat
  • a transgene is a DNA which is integrated into the genome of a cell from which a transgenic animal develops.
  • recombinant (transgenic) animal models can be engineered by introducing the coding portion of the genes encoding VEGF-E identified herein into the genome of animals of interest, using standard techniques for producing transgenic animals.
  • Animals that can serve as a target for transgenic manipulation include, without limitation, mice, rats, rabbits, guinea pigs, sheep, goats, pigs, and non-human primates, e . g ., baboons, chimpanzees and monkeys.
  • transgenic animals include those that carry the transgene only in part of their cells ("mosaic animals").
  • the transgene can be integrated either as a single transgene, or in concatamers, e . g ., head-to-head or head-to-tail tandems. Selective introduction of a transgene into a particular cell type is also possible by following, for example, the technique of Lasko et al., Proc. Natl. Acad. Sci. USA, 89: 6232-636 (1992 ).
  • the expression of the transgene in transgenic animals can be monitored by standard techniques. For example, Southern blot analysis or PCR amplification can be used to verify the integration of the transgene.
  • the level of mRNA expression can then be analyzed using techniques such as in situ hybridization, Northern blot analysis, PCR, or immunocytochemistry. The animals are further examined for signs of tumor or cancer development.
  • "knock-out" animals can be constructed that have a defective or altered gene encoding a VEGF-E polypeptide identified herein, as a result of homologous recombination between the endogenous gene encoding the VEGF-E polypeptide and altered genomic DNA encoding the same polypeptide introduced into an embryonic cell of the animal.
  • cDNA encoding a particular VEGF-E polypeptide can be used to clone genomic DNA encoding that polypeptide in accordance with established techniques.
  • a portion of the genomic DNA encoding a particular VEGF-E polypeptide can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration.
  • SCC feline oral squamous cell carcinoma
  • Feline oral SCC is a highly invasive, malignant tumor that is the most common oral malignancy of cats, accounting for over 60% of the oral tumors reported in this species. It rarely metastasizes to distant sites, although this low incidence of metastasis may merely be a reflection of the short survival times for cats with this tumor.
  • These tumors are usually not amenable to surgery, primarily because of the anatomy of the feline oral cavity. At present, there is no effective treatment for this tumor.
  • each cat Prior to entry into the study, each cat undergoes complete clinical examination and biopsy, and is scanned by computed tomography (CT). Cats diagnosed with sublingual oral squamous cell tumors are excluded from the study. The tongue can become paralyzed as a result of such tumor, and even if the treatment kills the tumor, the animals may not be able to feed themselves.
  • CT computed tomography
  • Each cat is treated repeatedly, over a longer period of time. Photographs of the tumors will be taken daily during the treatment period, and at each subsequent recheck.
  • CT scans and thoracic radiograms are evaluated every 8 weeks thereafter. The data are evaluated for differences in survival, response, and toxicity as compared to control groups. Positive response may require evidence of tumor regression, preferably with improvement of quality of life and/or increased life span.
  • cardiovascular, endothelial, and angiogenic assays herein can be verified by further studies, such as by determining mRNA expression in various human tissues.
  • exemplary antibodies include polyclonal, monoclonal, humanized, bispecific, and heteroconjugate antibodies, the preparation of which will be described hereinbelow.
  • Antibody binding studies may be carried out in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques (CRC Press, Inc., 1987), pp.147-158 .
  • ком ⁇ онентs rely on the ability of a labeled standard to compete with the test sample analyte for binding with a limited amount of antibody.
  • the amount of target protein in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies.
  • the antibodies preferably are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte that remain unbound.
  • the tissue sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin, for example.
  • suitable tumor cells include, for example, stable tumor cells lines such as the B104-1-1 cell line (stable NIH-3T3 cell line transfected with the neu protooncogene) and ras- transfected NIH-3T3 cells, which can be transfected with the desired gene and monitored for tumorigenic growth.
  • VEGF-E polypeptide herein and polypeptidyl agonists and antagonists may be employed in accordance with the present invention by expression of such polypeptides in vivo, which is often referred to as gene therapy.
  • a viral vector such as a retroviral vector includes at least one transcriptional promoter/enhancer or locus-defining element(s), or other elements that control gene expression by other means such as alternate splicing, nuclear RNA export, or post-translational modification of messenger.
  • a viral vector such as a retroviral vector includes a nucleic acid molecule that, when transcribed in the presence of a gene encoding VEGF-E polypeptide, is operably linked thereto and acts as a translation initiation sequence.
  • Such vector constructs also include a packaging signal, long terminal repeats (LTRs) or portions thereof, and positive and negative strand primer binding sites appropriate to the virus used (if these are not already present in the viral vector).
  • such vector typically includes a signal sequence for secretion of the VEGF-E polypeptide from a host cell in which it is placed.
  • the signal sequence for this purpose is a mammalian signal sequence, most preferably the native signal sequence for VEGF-E polypeptide.
  • the vector construct may also include a signal that directs polyadenylation, as well as one or more restriction sites and a translation termination sequence.
  • such vectors will typically include a 5' LTR, a tRNA binding site, a packaging signal, an origin of second-strand DNA synthesis, and a 3' LTR or a portion thereof.
  • Other vectors can be used that are non-viral, such as cationic lipids, polylysine, and dendrimers.
  • Suitable gene therapy and methods for making retroviral particles and structural proteins can be found in, e.g ., U.S. Pat. No. 5,681,746 .
  • This invention is also related to the use of the gene encoding the VEGF-E polypeptide as a diagnostic. Detection of a mutated form of the VEGF-E polypeptide will allow a diagnosis of a cardiovascular, endothelial, and angiogenic disease or a susceptibility to a cardiovascular, endothelial, and angiogenic disease, such as a tumor, since mutations in the VEGF-E polypeptide may cause tumors.
  • RNA or cDNA may also be used for the same purpose.
  • PCR primers complementary to the nucleic acid encoding the VEGF-E polypeptide can be used to identify and analyze VEGF-E polypeptide mutations.
  • deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype.
  • Point mutations can be identified by hybridizing amplified DNA to radiolabeled RNA encoding VEGF-E polypeptide, or alternatively, radiolabeled antisense DNA sequences encoding VEGF-E polypeptide. Perfectly matched sequences can be distinguished from mismatched duplexes by RNAse A digestion or by differences in melting temperatures.
  • Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNAse and S1 protection or the chemical cleavage method, for example, Cotton et al., Proc. Natl. Acad. Sci. USA, 85: 4397-4401 (1985 ).
  • the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNAse protection, chemical cleavage, direct DNA sequencing, or the use of restriction enzymes, e.g ., restriction fragment length polymorphisms (RFLP), and Southern blotting of genomic DNA.
  • restriction enzymes e.g ., restriction fragment length polymorphisms (RFLP), and Southern blotting of genomic DNA.
  • mutations can also be detected by in situ analysis.
  • VEGF-E polypeptide expression may be linked to vascular disease or neovascularization associated with tumor formation. If the VEGF-E polypeptide has a signal sequence and the mRNA is highly expressed in endothelial cells and to a lesser extent in smooth muscle cells, this indicates that the VEGF-E polypeptide is present in serum. Accordingly, an anti-VEGF-E polypeptide antibody could be used to diagnose vascular disease or neovascularization associated with tumor formation, since an altered level of this VEGF-E polypeptide may be indicative of such disorders.
  • Nucleotide sequences encoding a VEGF-E polypeptide can also be used to construct hybridization probes for mapping the gene which encodes that VEGF-E polypeptide and for the genetic analysis of individuals with genetic disorders.
  • the nucleotide sequence provided herein may be mapped to a chromosome and specific regions of a chromosome using known techniques, such as in situ hybridization, linkage analysis against known chromosomal markers, and hybridization screening with libraries.
  • chromosome identification the sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome. Moreover, there is a current need for identifying particular sites on the chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location.
  • the mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease. Briefly, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA.
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome.
  • sublocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner.
  • Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes, and preselection by hybridization to construct chromosome-specific cDNA libraries.
  • Fluorescence in situ hybridization (FISH) of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step.
  • This technique can be used with cDNA as short as 500 or 600 bases; however, clones larger than 2,000 bp have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection.
  • FISH requires use of the clones from which the gene encoding VEGF-E polypeptide was derived, and the longer the better. For example, 2,000 bp is good, 4,000 bp is better, and more than 4,000 is probably not necessary to get good results a reasonable percentage of the time.
  • Verma et al. Human Chromosomes: a Manual of Basic Techniques (Pergamon Press, New York, 1988 ).
  • Screening assays can be designed to find lead compounds that mimic the biological activity of a native VEGF-E or a receptor for VEGF-E. Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candidates. Small molecules contemplated include synthetic organic or inorganic compounds. The assays can be performed in a variety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays and cell based assays, which are well characterized in the art.
  • this invention encompasses methods of screening compounds to identify those that mimic the VEGF-E polypeptide (agonists) or prevent the effect of the VEGF-E polypeptide (antagonists).
  • Screening assays for antagonist drug candidates are designed to identify compounds that bind or complex with the VEGF-E polypeptides encoded by the genes identified herein, or otherwise interfere with the interaction of the encoded polypeptides with other cellular proteins.
  • Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candidates.
  • All assays for antagonists are common in that they call for contacting the drug candidate with a VEGF-E polypeptide encoded by a nucleic acid identified herein under conditions and for a time sufficient to allow these two components to interact.
  • the interaction is binding and the complex formed can be isolated or detected in the reaction mixture.
  • the VEGF-E polypeptide encoded by the gene identified herein or the drug candidate is immobilized on a solid phase, e.g ., on a microtiter plate, by covalent or non-covalent attachments.
  • Non-covalent attachment generally is accomplished by coating the solid surface with a solution of the VEGF-E polypeptide and drying.
  • an immobilized antibody e.g ., a monoclonal antibody, specific for the VEGF-E polypeptide to be immobilized can be used to anchor it to a solid surface.
  • the candidate compound interacts with but does not bind to a particular VEGF-E polypeptide encoded by a gene identified herein, its interaction with that polypeptide can be assayed by methods well known for detecting protein-protein interactions.
  • assays include traditional approaches, such as, e.g ., cross-linking, co-immunoprecipitation, and co-purification through gradients or chromatographic columns.
  • protein-protein interactions can be monitored by using a yeast-based genetic system described by Fields and co-workers ( Fields and Song, Nature (London), 340: 245-246 (1989 ); Chien et al., Proc. Natl. Acad. Sci.
  • yeast GAL4 consist of two physically discrete modular domains, one acting as the DNA-binding domain, the other one functioning as the transcription-activation domain.
  • the yeast expression system described in the foregoing publications (generally referred to as the "two-hybrid system") takes advantage of this property, and employs two hybrid proteins, one in which the target protein is fused to the DNA-binding domain of GAL4, and another, in which candidate activating proteins are fused to the activation domain.
  • GAL1- lac Z reporter gene under control of a GAL4-activated promoter depends on reconstitution of GAL4 activity via protein-protein interaction. Colonies containing interacting polypeptides are detected with a chromogenic substrate for ⁇ -galactosidase.
  • a complete kit (MATCHMAKER TM ) for identifying protein-protein interactions between two specific proteins using the two-hybrid technique is commercially available from Clontech. This system can also be extended to map protein domains involved in specific protein interactions as well as to pinpoint amino acid residues that are crucial for these interactions.
  • the assay described above is performed; however, in this assay the VEGF-E polypeptide is added along with the compound to be screened and the ability of the compound to inhibit 3- (H)thymidine incorporation in the presence of the VEGF-E polypeptide indicates that the compound is an antagonist to the VEGF-E polypeptide.
  • antagonists may be detected by combining the VEGF-E polypeptide and a potential antagonist with membrane-bound VEGF-E polypeptide receptors or recombinant receptors under appropriate conditions for a competitive inhibition assay.
  • labeled VEGF-E polypeptide can be photoaffinity-linked with cell membrane or extract preparations that express the receptor molecule. Cross-linked material is resolved by PAGE and exposed to X-ray film. The labeled complex containing the receptor can be excised, resolved into peptide fragments, and subjected to protein micro-sequencing. The amino acid sequence obtained from micro- sequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the gene encoding the putative receptor.
  • compositions useful in the treatment of cardiovascular, endothelial, and angiogenic disorders include, without limitation, antibodies, small organic and inorganic molecules, peptides, phosphopeptides, antisense and ribozyme molecules, triple-helix molecules, etc., that inhibit the expression and/or activity of the target gene product.
  • potential antagonists include an oligonucleotide that binds to the VEGF-E polypeptide, (poly)peptide immunoglobulin fusions, and, in particular, antibodies including, without limitation, poly- and monoclonal antibodies and antibody fragments, single-chain antibodies, anti-idiotypic antibodies, and chimeric or humanized versions of such antibodies or fragments, as well as human antibodies and antibody fragments.
  • a potential antagonist may be a closely related protein, for example, a mutated form of the VEGF-E polypeptide that recognizes the receptor but imparts no effect, thereby competitively inhibiting the action of the VEGF-E polypeptide.
  • VEGF-E polypeptide antagonist is an antisense RNA or DNA construct prepared using antisense technology, where, e.g., an antisense RNA or DNA molecule acts to block directly the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation.
  • Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA.
  • the 5' coding portion of the polynucleotide sequence which encodes the mature VEGF-E polypeptides herein, is used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length.
  • a DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple helix - see Lee et al., Nucl. Acids Res., 6: 3073 (1979 ); Cooney et al., Science, 241: 456 (1988 ); Dervan et al., Science, 251: 1360 (1991 )), thereby preventing transcription and the production of the VEGF-E polypeptide.
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the VEGF-E polypeptide (antisense - Okano, Neurochem., 56: 560 (1991 ); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression (CRC Press: Boca Raton, FL, 1988 ).
  • the oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the VEGF-E polypeptide.
  • antisense DNA is used, oligodeoxyribonucleotides derived from the translation-initiation site, e.g ., between about - 10 and +10 positions of the target gene nucleotide sequence, are preferred.
  • Potential antagonists include small molecules that bind to the active site, the receptor binding site, or growth factor or other relevant binding site of the VEGF-E polypeptide, thereby blocking the normal biological activity of the VEGF-E polypeptide.
  • small molecules include, but are not limited to, small peptides or peptide-like molecules, preferably soluble peptides, and synthetic non-peptidyl organic or inorganic compounds.
  • Their therapeutic utility could include diseases of the arteries, capillaries, veins, and/or lymphatics.
  • treatments hereunder include treating muscle wasting disease, treating osteoporosis, aiding in implant fixation to stimulate the growth of cells around the implant and therefore facilitate its attachment to its intended site, increasing IGF stability in tissues or in serum, if applicable, and increasing binding to the IGF receptor (since IGF has been shown in vitro to enhance human marrow erythroid and granulocytic progenitor cell growth).
  • VEGF-E polypeptides or agonists or antagonists thereto may also be employed to stimulate erythropoiesis or granulopoiesis, to stimulate wound healing or tissue regeneration and associated therapies concerned with re-growth of tissue, such as connective tissue, skin, bone, cartilage, muscle, lung, or kidney, to promote angiogenesis, to stimulate or inhibit migration of endothelial cells, and to proliferate the growth of vascular smooth muscle and endothelial cell production.
  • tissue such as connective tissue, skin, bone, cartilage, muscle, lung, or kidney
  • angiogenesis to stimulate or inhibit migration of endothelial cells
  • the increase in angiogenesis mediated by VEGF-E polypeptide or antagonist would be beneficial to ischemic tissues and to collateral coronary development in the heart subsequent to coronary stenosis.
  • Antagonists are used to inhibit the action of such polypeptides, for example, to limit the production of excess connective tissue during wound healing or pulmonary fibrosis if the VEGF-E polypeptide promotes such production. This would include treatment of acute myocardial infarction and heart failure.
  • the present invention concerns means for the treatment of cardiac hypertrophy, regardless of the underlying cause, by administering a therapeutically effective dose of VEGF-E polypeptide, or agonist or antagonist thereto.
  • the objective is the treatment of human patients
  • the VEGF-E polypeptide preferably is recombinant human VEGF-E polypeptide (rhVEGF-E polypeptide).
  • the treatment for cardiac hypertrophy can be performed at any of its various stages, which may result from a variety of diverse pathologic conditions, including myocardial infarction, hypertension, hypertrophic cardiomyopathy, and valvular regurgitation.
  • the treatment extends to all stages of the progression of cardiac hypertrophy, with or without structural damage of the heart muscle, regardless of the underlying cardiac disorder.
  • the decision of whether to use the molecule itself or an agonist thereof for any particular indication, as opposed to an antagonist to the molecule, would depend mainly on whether the molecule herein promotes cardiovascularization, genesis of endothelial cells, or angiogenesis or inhibits these conditions. For example, if the molecule promotes angiogenesis, an antagonist thereof would be useful for treatment of disorders where it is desired to limit or prevent angiogenesis.
  • vascular tumors such as haemangioma, tumor angiogenesis, neovascularization in the retina, choroid, or cornea, associated with diabetic retinopathy or premature infant retinopathy or macular degeneration and proliferative vitreoretinopathy, rheumatoid arthritis, Crohn's disease, atherosclerosis, ovarian hyperstimulation, psoriasis, endometriosis associated with neovascularization, restenosis subsequent to balloon angioplasty, scar tissue overproduction, for example, that seen in a keloid that forms after surgery, fibrosis after myocardial infarction, or fibrotic lesions associated with pulmonary fibrosis.
  • vascular tumors such as haemangioma, tumor angiogenesis, neovascularization in the retina, choroid, or cornea, associated with diabetic retinopathy or premature infant retinopathy or macular degeneration and proliferative vitreoretinopathy, rheumato
  • the molecule inhibits angiogenesis, it would be expected to be used directly for treatment of the above conditions.
  • angiogenesis it would be used itself (or an agonist thereof) for indications where angiogenesis is desired such as peripheral vascular disease, hypertension, inflammatory vasculitides, Reynaud's disease and Reynaud's phenomenon, aneurysms, arterial restenosis, thrombophlebitis, lymphangitis, lymphedema, wound healing and tissue repair, ischemia reperfusion injury, angina, myocardial infarctions such as acute myocardial infarctions, chronic heart conditions, heart failure such as congestive heart failure, and osteoporosis.
  • Atherosclerosis is a disease characterized by accumulation of plaques of intimal thickening in arteries, due to accumulation of lipids, proliferation of smooth muscle cells, and formation of fibrous tissue within the arterial wall.
  • the disease can affect large, medium, and small arteries in any organ. Changes in endothelial and vascular smooth muscle cell function are known to play an important role in modulating the accumulation and regression of these plaques.
  • Hypertension is characterized by raised vascular pressure in the systemic arterial, pulmonary arterial, or portal venous systems. Elevated pressure may result from or result in impaired endothelial function and/or vascular disease.
  • Inflammatory vasculitides include giant cell arteritis, Takayasu's arteritis, polyarteritis nodosa (including the microangiopathic form), Kawasaki's disease, microscopic polyangiitis, Wegener's granulomatosis, and a variety of infectious-related vascular disorders (including Henoch-Schonlein prupura). Altered endothelial cell function has been shown to be important in these diseases.
  • Thrombophlebitis and lymphangitis are inflammatory disorders of veins and lymphatics, respectively, that may result from, and/or in, altered endothelial cell function.
  • lymphedema is a condition involving impaired lymphatic vessels resulting from endothelial cell function.
  • lymphangiomas are benign tumors of the lymphatic system that are congenital, often cystic, malformations of the lymphatics that usually occur in newborns. Cystic tumors tend to grow into the adjacent tissue. Cystic tumors usually occur in the cervical and axillary region. They can also occur in the soft tissue of the extremities. The main symptoms are dilated, sometimes reticular, structured lymphatics and lymphocysts surrounded by connective tissue. Lymphangiomas are assumed to be caused by improperly connected embryonic lymphatics or their deficiency. The result is impaired local lymph drainage. Griener et al., Lymphology, 4: 140-144 (1971 ).
  • AMD Age-related macular degeneration
  • AMD Age-related macular degeneration
  • the exudative form of AMD is characterized by choroidal neovascularization and retinal pigment epithelial cell detachment. Because choroidal neovascularization is associated with a dramatic worsening in prognosis, the VEGF-E polypeptides or antagonist thereto is expected to be useful in reducing the severity of AMD.
  • VEGF-E polypeptides herein or their antagonists are also a targeted use for the VEGF-E polypeptides herein or their antagonists. Formation and regression of new blood vessels is essential for tissue healing and repair. This category includes bone, cartilage, tendon, ligament, and/or nerve tissue growth or regeneration, as well as wound healing and tissue repair and replacement, and in the treatment of burns, incisions, and ulcers.
  • a VEGF-E polypeptide or antagonist thereof that induces cartilage and/or bone growth in circumstances where bone is not normally formed has application in the healing of bone fractures and cartilage damage or defects in humans and other animals.
  • Such a preparation employing a VEGF-E polypeptide or antagonist thereof may have prophylactic use in closed as well as open fracture reduction and also in the improved fixation of artificial joints.
  • De novo bone formation induced by an osteogenic agent contributes to the repair of congenital, trauma-induced, or oncologic, resection-induced craniofacial defects, and also is useful in cosmetic plastic surgery.
  • VEGF-E polypeptides or antagonists thereto may also be useful to promote better or faster closure of non-healing wounds, including without limitation pressure ulcers, ulcers associated with vascular insufficiency, surgical and traumatic wounds, and the like.
  • a VEGF-E polypeptide or antagonist thereto may also exhibit activity for generation or regeneration of other tissues, such as organs (including, for example, pancreas, liver, intestine, kidney, skin, or endothelium), muscle (smooth, skeletal, or cardiac), and vascular (including vascular endothelium) tissue, or for promoting the growth of cells comprising such tissues.
  • organs including, for example, pancreas, liver, intestine, kidney, skin, or endothelium
  • muscle smooth, skeletal, or cardiac
  • vascular including vascular endothelium
  • a VEGF-E polypeptide herein or antagonist thereto may also be useful for gut protection or regeneration and treatment of lung or liver fibrosis, reperfusion injury in various tissues, and conditions resulting from systemic cytokine damage. Also, the VEGF-E polypeptide or antagonist thereto may be useful for promoting or inhibiting differentiation of tissues described above from precursor tissues or cells, or for inhibiting the growth of tissues described above.
  • a VEGF-E polypeptide or antagonist thereto may also be used in the treatment of periodontal diseases and in other tooth-repair processes. Such agents may provide an environment to attract bone-forming cells, stimulate growth of bone-forming cells, or induce differentiation of progenitors of bone-forming cells.
  • a VEGF-E polypeptide herein or an antagonist thereto may also be useful in the treatment of osteoporosis or osteoarthritis, such as through stimulation of bone and/or cartilage repair or by blocking inflammation or processes of tissue destruction (collagenase activity, osteoclast activity, etc.) mediated by inflammatory processes, since blood vessels play an important role in the regulation of bone turnover and growth.
  • tissue regeneration activity that may be attributable to the VEGF-E polypeptide herein or antagonist thereto is tendon/ligament formation.
  • a protein that induces tendon/ligament-like tissue or other tissue formation in circumstances where such tissue is not normally formed has application in the healing of tendon or ligament tears, deformities, and other tendon or ligament defects in humans and other animals.
  • Such a preparation may have prophylactic use in preventing damage to tendon or ligament tissue, as well as use in the improved fixation of tendon or ligament to bone or other tissues, and in repairing defects to tendon or ligament tissue.
  • compositions herein may provide an environment to attract tendon- or ligament-forming cells, stimulate growth of tendon- or ligament-forming cells, induce differentiation of progenitors of tendon- or ligament-forming cells, or induce growth of tendon/ligament cells or progenitors ex vivo for return in vivo to effect tissue repair.
  • the compositions herein may also be useful in the treatment of tendinitis, carpal tunnel syndrome, and other tendon or ligament defects.
  • the compositions may also include an appropriate matrix and/or sequestering agent as a carrier as is well known in the art.
  • the VEGF-E polypeptide or its antagonist may also be useful for proliferation of neural cells and for regeneration of nerve and brain tissue, i.e., for the treatment of central and peripheral nervous system disease and neuropathies, as well as mechanical and traumatic disorders, that involve degeneration, death, or trauma to neural cells or nerve tissue. More specifically, a VEGF-E polypeptide or its antagonist may be used in the treatment of diseases of the peripheral nervous system, such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome.
  • diseases of the peripheral nervous system such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome.
  • VEGF-E polypeptide herein or antagonist thereto.
  • Ischemia-reperfusion injury is another indication. Endothelial cell dysfunction may be important in both the initiation of, and in regulation of the sequelae of events that occur following ischemia-reperfusion injury.
  • Rheumatoid arthritis is a further indication.
  • Blood vessel growth and targeting of inflammatory cells through the vasculature is an important component in the pathogenesis of rheumatoid and sero-negative forms of arthritis.
  • VEGF-E polypeptide or its antagonist may also be administered prophylactically to patients with cardiac hypertrophy, to prevent the progression of the condition, and avoid sudden death, including death of asymptomatic patients.
  • Such preventative therapy is particularly warranted in the case of patients diagnosed with massive left ventricular cardiac hypertrophy (a maximal wall thickness of 35 mm or more in adults, or a comparable value in children), or in instances when the hemodynamic burden on the heart is particularly strong.
  • VEGF-E polypeptide or its antagonist may also be useful in the management of atrial fibrillation, which develops in a substantial portion of patients diagnosed with hypertrophic cardiomyopathy.
  • VEGF-E polypeptides or agonists or antagonists thereof described herein which are shown to alter or impact endothelial cell function, proliferation, and/or form, are likely to play an important role in the etiology and pathogenesis of many or all of the disorders noted above, and as such can serve as therapeutic targets to augment or inhibit these processes or for vascular-related drug targeting in these disorders.
  • the molecules herein and agonists and antagonists thereto are pharmaceutically useful as a prophylactic and therapeutic agent for various disorders and diseases as set forth above.
  • the VEGF-E of the present invention can be formulated according to known methods to prepare pharmaceutically-useful compositions, whereby the VEGF-E hereof is combined in admixture with a pharmaceutically acceptable carrier vehicle.
  • Suitable carrier vehicles and their formulation, inclusive of other human proteins, e.g., human serum albumin, are described, for example, in Remington's Pharmaceutical Sciences, 16th ed., 1980, Mack Publishing Co., edited by Oslo et al .
  • the VEGF-E herein may be administered parenterally to subjects suffering from cardiovascular diseases or conditions, or by other methods that ensure its delivery to the bloodstream in an effective form.
  • compositions particularly well suited for the clinical administration of VEGF-E hereof employed in the practice of the present invention include, for example, sterile aqueous solutions, or sterile hydratable powders such as lyophilized protein. It is generally desirable to include further in the formulation an appropriate amount of a pharmaceutically acceptable salt, generally in an amount sufficient to render the formulation isotonic.
  • a pH regulator such as arginine base, and phosphoric acid, are also typically included in sufficient quantities to maintain an appropriate pH, generally from 5.5 to 7.5.
  • further agents such as glycerol. In this manner, variant VEGF-E formulations are rendered appropriate for parenteral administration, and, in particular, intravenous administration.
  • compositions of the VEGF-E polypeptides or agonists or antagonists are prepared for storage by mixing the desired molecule having the appropriate degree of purity with optional pharmaceutically acceptable carriers, excipients, or stabilizers ( Remington's Pharmaceutical Sciences, 16th edition, Oslo, A. ed. (1980 )), in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • Such carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, and polyethylene glycol.
  • buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrroli
  • Another formulation comprises incorporating a VEGF-E polypeptide or antagonist thereof into formed articles.
  • Such articles can be used in modulating endothelial cell growth and angiogenesis.
  • tumor invasion and metastasis may be modulated with these articles.
  • the VEGF-E to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e.g., 0.2 micron membranes).
  • the VEGF-E ordinarily will be stored in lyophilized form or as an aqueous solution if it is highly stable to thermal and oxidative denaturation.
  • the pH of the VEGF-E preparations typically will be about from 6 to 8, although higher or lower pH values may also be appropriate in certain instances. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of salts of the VEGF-E.
  • An isotonifier may be present to ensure isotonicity of a liquid composition of the VEGF-E polypeptide or antagonist thereto, and includes polyhydric sugar alcohols, preferably trihydric or higher sugar alcohols, such as glycerin, erythritol, arabitol, xylitol, sorbitol, and mannitol. These sugar alcohols can be used alone or in combination. Alternatively, sodium chloride or other appropriate inorganic salts may be used to render the solutions isotonic.
  • the buffer may, for example, be an acetate, citrate, succinate, or phosphate buffer depending on the pH desired.
  • the pH of one type of liquid formulation of this invention is buffered in the range of about 4 to 8, preferably about physiological pH.
  • the preservatives phenol, benzyl alcohol and benzethonium halides, e.g., chloride, are known antimicrobial agents that may be employed.
  • Therapeutic VEGF-E polypeptide compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • the formulations are preferably administered as repeated intravenous (i.v.), subcutaneous (s.c.), or intramuscular (i.m.) injections, or as aerosol formulations suitable for intranasal or intrapulmonary delivery (for intrapulmonary delivery see, e . g ., EP 257, 956 ).
  • VEGF-E polypeptide can also be administered in the form of sustained-released preparations.
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the protein, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels ( e . g ., poly(2-hydroxyethyl-methacrylate) as described by Langer et al., J. Biomed. Mater. Res., 15: 167-277 (1981 ) and Langer, Chem.
  • VEGF-E polypeptide compositions also include liposomally entrapped VEGF-E polypeptide.
  • Liposomes containing VEGF-E polypeptide are prepared by methods known per se : DE 3,218,121 ; Epstein et al., Proc. Natl. Acad. Sci. USA, 82: 3688-3692 (1985 ); Hwang et al., Proc.
  • the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. % cholesterol, the selected proportion being adjusted for the optimal therapy.
  • VEGF-E polypeptide or antagonist thereto will, of course, vary depending on such factors as the pathological condition to be treated (including prevention), the method of administration, the type of compound being used for treatment, any co-therapy involved, the patient's age, weight, general medical condition, medical history, etc., and its determination is well within the skill of a practicing physician. Accordingly, it will be necessary for the therapist to titer the dosage and modify the route of administration as required to obtain the maximal therapeutic effect. If the VEGF-E polypeptide has a narrow host range, for the treatment of human patients formulations comprising human VEGF-E polypeptide, more preferably native-sequence human VEGF-E polypeptide, are preferred.
  • VEGF-E polypeptide The clinician will administer VEGF-E polypeptide until a dosage is reached that achieves the desired effect for treatment of the condition in question. For example, if the objective is the treatment of CHF, the amount would be one that inhibits the progressive cardiac hypertrophy associated with this condition. The progress of this therapy is easily monitored by echo cardiography. Similarly, in patients with hypertrophic cardiomyopathy, VEGF-E polypeptide can be administered on an empirical basis.
  • the effective dose generally is within the range of from about 0.001 to about 1.0 mg/kg, more preferably about 0.01-1 mg/kg, most preferably about 0.01-0.1 mg/kg.
  • VEGF-E polypeptide for non-oral use in treating human adult hypertension, it is advantageous to administer VEGF-E polypeptide in the form of an injection at about 0.01 to 50 mg, preferably about 0.05 to 20 mg, most preferably 1 to 20 mg, per kg body weight, 1 to 3 times daily by intravenous injection.
  • a molecule based on the VEGF-E polypeptide is preferably administered at about 5 mg to 1 g, preferably about 10 to 100 mg, per kg body weight, 1 to 3 times daily.
  • endotoxin contamination should be kept minimally at a safe level, for example, less than 0.5 ng/mg protein.
  • the formulations preferably meet sterility, pyrogenicity, general safety, and purity as required by FDA Office and Biologics standards.
  • the dosage regimen of a pharmaceutical composition containing VEGF-E polypeptide to be used in tissue regeneration will be determined by the attending physician considering various factors that modify the action of the polypeptides, e . g ., amount of tissue weight desired to be formed, the site of damage, the condition of the damaged tissue, the size of a wound, type of damaged tissue ( e . g ., bone), the patient's age, sex, and diet, the severity of any infection, time of administration, and other clinical factors.
  • the dosage may vary with the type of matrix used in the reconstitution and with inclusion of other proteins in the pharmaceutical composition. For example, the addition of other known growth factors, such as IGF-I, to the final composition may also affect the dosage. Progress can be monitored by periodic assessment of tissue/bone growth and/or repair, for example, X-rays, histomorphometric determinations, and tetracycline labeling.
  • VEGF-E polypeptide or antagonist or agonist administration is in accord with known methods, e.g., by injection or infusion by intravenous, intramuscular, intracerebral, intraperitoneal, intracerobrospinal, subcutaneous, intraocular, intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes, or by sustained-release systems as noted below.
  • the VEGF-E polypeptide or antagonists thereof also are suitably administered by intratumoral, peritumoral, intralesional, or perilesional routes, to exert local as well as systemic therapeutic effects.
  • the intraperitoneal route is expected to be particularly useful, for example, in the treatment of ovarian tumors.
  • a peptide or small molecule is employed as an antagonist or agonist, it is preferably administered orally or non-orally in the form of a liquid or solid to mammals.
  • Examples of pharmacologically acceptable salts of molecules that form salts and are useful hereunder include alkali metal salts (e . g ., sodium salt, potassium salt), alkaline earth metal salts (e . g ., calcium salt, magnesium salt), ammonium salts, organic base salts ( e . g ., pyridine salt, triethylamine salt), inorganic acid salts ( e . g ., hydrochloride, sulfate, nitrate), and salts of organic acid ( e . g ., acetate, oxalate, p-toluenesulfonate).
  • alkali metal salts e . g ., sodium salt, potassium salt
  • alkaline earth metal salts e . g ., calcium salt, magnesium salt
  • ammonium salts e e g ., organic base salts ( e . g ., pyridine salt, triethy
  • the therapeutic method includes administering the composition topically, systemically, or locally as an implant or device.
  • the therapeutic composition for use is in a pyrogen-free, physiologically acceptable form.
  • the composition may desirably be encapsulated or injected in a viscous form for delivery to the site of bone, cartilage, or tissue damage.
  • Topical administration may be suitable for wound healing and tissue repair.
  • the composition would include a matrix capable of delivering the protein-containing composition to the site of bone and/or cartilage damage, providing a structure for the developing bone and cartilage and preferably capable of being resorbed into the body.
  • Such matrices may be formed of materials presently in use for other implanted medical applications.
  • One specific embodiment is a 50:50 (mole weight) copolymer of lactic acid and glycolic acid in the form of porous particles having diameters ranging from 150 to 800 microns.
  • a sequestering agent such as carboxymethyl cellulose or autologous blood clot, to prevent the polypeptide compositions from disassociating from the matrix.
  • One suitable family of sequestering agents is cellulosic materials such as alkylcelluloses (including hydroxyalkylcelluloses), including methylcellulose, ethylcellulose, hydoxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, and carboxymethylcellulose, one preferred being cationic salts of carboxymethylcellulose (CMC).
  • alkylcelluloses including hydroxyalkylcelluloses
  • methylcellulose ethylcellulose, hydoxyethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, and carboxymethylcellulose, one preferred being cationic salts of carboxymethylcellulose (CMC).
  • CMC carboxymethylcellulose
  • Other preferred sequestering agents include hyaluronic acid, sodium alginate, poly(ethylene glycol), polyoxyethylene oxide, carboxyvinyl polymer, and poly(vinyl alcohol).
  • the amount of sequestering agent useful herein is 0.5-20 wt%, preferably 1-10 wt%, based on total formulation weight, which represents the amount necessary to prevent desorption of the polypeptide (or its antagonist) from the polymer matrix and to provide appropriate handling of the composition, yet not so much that the progenitor cells are prevented from infiltrating the matrix, thereby providing the polypeptide (or its antagonist) the opportunity to assist the osteogenic activity of the progenitor cells.
  • VEGF-E vascular endothelial growth factor
  • the VEGF-E formulated in a liquid composition may be mixed with an effective amount of a water-soluble polysaccharide or synthetic polymer such as polyethylene glycol to form a gel of the proper viscosity to be applied topically.
  • a water-soluble polysaccharide or synthetic polymer such as polyethylene glycol
  • the polysaccharide that may be used includes, for example, cellulose derivatives such as etherified cellulose derivatives, including alkyl celluloses, hydroxyalkyl celluloses, and alkylhydroxyalkyl celluloses, for example, methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxypropyl methylcellulose, and hydroxypropyl cellulose; starch and fractionated starch; agar; alginic acid and alginates; gum arabic; pullullan; agarose; carrageenan; dextrans; dextrins; fructans; inulin; mannans; xylans; arabinans; chitosans; glycogens; glucans; and synthetic biopolymers; as well as gums such as xanthan gum; guar gum; locust bean gum; gum arabic; tragacanth gum; and karaya gum; and derivatives and mixtures thereof.
  • the preferred gelling agent herein is one that is in
  • the polysaccharide is an etherified cellulose derivative, more preferably one that is well defined, purified, and listed in USP, e.g., methylcellulose and the hydroxyalkyl cellulose derivatives, such as hydroxypropyl cellulose, hydroxyethyl cellulose, and hydroxypropyl methylcellulose. Most preferred herein is methylcellulose.
  • the polyethylene glycol useful for gelling is typically a mixture of low- and high-molecular-weight polyethylene glycols to obtain the proper viscosity.
  • a mixture of a polyethylene glycol of molecular weight 400-600 with one of molecular weight 1500 would be effective for this purpose when mixed in the proper ratio to obtain a paste.
  • water soluble as applied to the polysaccharides and polyethylene glycols is meant to include colloidal solutions and dispersions.
  • solubility of the cellulose derivatives is determined by the degree of substitution of ether groups, and the stabilizing derivatives useful herein should have a sufficient quantity of such ether groups per anhydroglucose unit in the cellulose chain to render the derivatives water soluble.
  • a degree of ether substitution of at least 0.35 ether groups per anhydroglucose unit is generally sufficient.
  • the cellulose derivatives may be in the form of alkali metal salts, for example, the Li, Na, K, or Cs salts.
  • methylcellulose is employed in the gel, preferably it comprises about 2-5%, more preferably about 3%, of the gel and the VEGF-E is present in an amount of about 300-1000 mg per ml of gel.
  • VEGF-E polypeptide or an agonist or antagonist thereof in preventing or treating the disorder in question may be improved by administering the active agent serially or in combination with another agent that is effective for those purposes, either in the same composition or as separate compositions.
  • VEGF-E therapy with other novel or conventional therapies (e.g., growth factors such as VEGF, aFGF, bFGF, PDGF, IGF, NGF, anabolic steroids, EGF or TGF-alpha) for enhancing the activity of any of the growth factors, including VEGF-E, in promoting cell proliferation, survival, differentiation, and repair.
  • cotreatment drugs be included per se in the compositions of this invention, although this will be convenient where such drugs are proteinaceous.
  • Such admixtures are suitably administered in the same manner and for the same purposes as the VEGF-E used alone.
  • VEGF-E polypeptide therapy can be combined with the administration of inhibitors of known cardiac myocyte hypertrophy factors, e . g ., inhibitors of ⁇ -adrenergic agonists such as phenylephrine; endothelin-1 inhibitors such as BOSENTAN TM and MOXONODIN TM ; inhibitors to CT-1 ( US Pat. No. 5,679,545 ); inhibitors to LIF; ACE inhibitors; des-aspartate-angiotensin I inhibitors ( U.S. Pat. No. 5,773,415 ), and angiotensin II inhibitors.
  • inhibitors of known cardiac myocyte hypertrophy factors e . g ., inhibitors of ⁇ -adrenergic agonists such as phenylephrine; endothelin-1 inhibitors such as BOSENTAN TM and MOXONODIN TM ; inhibitors to CT-1 ( US Pat. No. 5,679,545 ); inhibitors to LIF;
  • VEGF-E polypeptide can be administered in combination with ⁇ -adrenergic receptor blocking agents, e . g ., propranolol, timolol, tertalolol, carteolol, nadolol, betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, or carvedilol; ACE inhibitors, e .
  • ⁇ -adrenergic receptor blocking agents e . g ., propranolol, timolol, tertalolol, carteolol, nadolol, betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, or carvedilol
  • ACE inhibitors e .
  • quinapril e.g ., quinapril, captopril, enalapril, ramipril, benazepril, fosinopril, or lisinopril
  • diuretics e . g ., chorothiazide, hydrochlorothiazide, hydroflumethazide, methylchlothiazide, benzthiazide, dichlorphenamide, acetazolamide, or indapamide
  • calcium channel blockers e . g ., diltiazem, nifedipine, verapamil, or nicardipine.
  • compositions comprising the therapeutic agents identified herein by their generic VEGF-Es are commercially available, and are to be administered following the manufacturers' instructions for dosage, administration, adverse effects, contraindications, etc. See, e . g ., Physicians' Desk Reference (Medical Economics Data Production Co.: Montvale, N.J., 1997), 51th Editi on.
  • Preferred candidates for combination therapy in the treatment of hypertrophic cardiomyopathy are ⁇ -adrenergic-blocking drugs (e . g ., propranolol, timolol, tertalolol, carteolol, nadolol, betaxolol, penbutolol, acetobutolol, atenolol, metoprolol, or carvedilol), verapamil, difedipine, or diltiazem.
  • Treatment of hypertrophy associated with high blood pressure may require the use of antihypertensive drug therapy, using calcium channel blockers, e .
  • diltiazem nifedipine, verapamil, or nicardipine
  • ⁇ -adrenergic blocking agents diuretics, e . g ., chorothiazide, hydrochlorothiazide, hydroflumethazide, methylchlothiazide, benzthiazide, dichlorphenamide, acetazolamide, or indapamide
  • ACE-inhibitors e . g ., quinapril, captopril, enalapril, ramipril, benazepril, fosinopril, or lisinopril.
  • VEGF-E polypeptides or their antagonists may be combined with other agents beneficial to the treatment of the bone and/or cartilage defect, wound, or tissue in question.
  • agents include various growth factors such as EGF, PDGF, TGF- ⁇ or TGF- ⁇ , IGF, FGF, and CTGF.
  • VEGF-E polypeptides or their antagonists used to treat cancer may be combined with cytotoxic, chemotherapeutic, or growth-inhibitory agents as identified above.
  • the VEGF-E polypeptide or antagonist thereof is suitably administered serially or in combination with radiological treatments, whether involving irradiation or administration of radioactive substances.
  • the effective amounts of the therapeutic agents administered in combination with VEGF-E polypeptide or antagonist thereof will be at the physician's or veterinarian's discretion. Dosage administration and adjustment is done to achieve maximal management of the conditions to be treated. For example, for treating hypertension, these amounts ideally take into account use of diuretics or digitalis, and conditions such as hyper- or hypotension, renal impairment, etc.
  • the dose will additionally depend on such factors as the type of the therapeutic agent to be used and the specific patient being treated. Typically, the amount employed will be the same dose as that used, if the given therapeutic agent is administered without VEGF-E polypeptide.
  • a useful molar ratio of VEGF-E to secondary growth factors is typically 1:0.1-10, with about equimolar amounts being preferred.
  • An article of manufacture such as a kit containing VEGF-E polypeptide or antagonists thereof useful for the diagnosis or treatment of the disorders described above comprises at least a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition that is effective for diagnosing or treating the condition and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agent in the composition is the VEGF-E polypeptide or an agonist or antagonist thereto.
  • immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor.
  • adjuvants which may be employed include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
  • the immunization protocol may be selected by one skilled in the art without undue experimentation.
  • the antibodies are used for treating cancer, it may be desirable also to administer antibodies against other tumor-associated antigens, such as antibodies that bind to one or more of the ErbB2, EGFR, ErbB3, ErbB4, or VEGF receptor(s). These also include the agents set forth above.
  • the antibody is suitably administered serially or in combination with radiological treatments, whether involving irradiation or administration of radioactive substances.
  • two or more antibodies binding the same or two or more different antigens disclosed herein may be co-administered to the patient.
  • the antibodies herein are co-administered with a growth-inhibitory agent.
  • the growth-inhibitory agent may be administered first, followed by an antibody of the present invention.
  • simultaneous administration or administration of the antibody of the present invention first is also contemplated.
  • Suitable dosages for the growth-inhibitory agent are those presently used and may be lowered due to the combined action (synergy) of the growth-inhibitory agent and the antibody herein.
  • vascularization of tumors is attacked in combination therapy.
  • the anti-VEGF-E polypeptide and another antibody e . g ., anti-VEGF
  • the anti-VEGF-E polypeptide and another antibody are administered to tumor-bearing patients at therapeutically effective doses as determined, for example, by observing necrosis of the tumor or its metastatic foci, if any. This therapy is continued until such time as no further beneficial effect is observed or clinical examination shows no trace of the tumor or any metastatic foci.
  • auxiliary agents will vary in their effectiveness, it is desirable to compare their impact on the tumor by matrix screening in conventional fashion.
  • the administration of anti-VEGF-E polypeptide antibody and TNF is repeated until the desired clinical effect is achieved.
  • the anti-VEGF-E polypeptide antibody is administered together with TNF and, optionally, auxiliary agent(s).
  • the therapeutic agents described herein are administered to the isolated tumor or organ.
  • a FGF or PDGF antagonist such as an anti-FGF or an anti-PDGF neutralizing antibody, is administered to the patient in conjunction with the anti-VEGF-E polypeptide antibody.
  • Treatment with anti-VEGF-E polypeptide antibodies preferably may be suspended during periods of wound healing or desirable neovascularization.
  • an antibody herein will depend on the type of disorder to be treated, as defined above, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments.
  • An article of manufacture containing a container with the antibody and a label is also provided. Such articles are described above, wherein the active agent is an anti-VEGF-E antibody.
  • antibodies can be used qualitatively or quantitatively to detect the expression of genes including the gene encoding the VEGF-E polypeptide.
  • the antibody preferably is equipped with a detectable, e.g., fluorescent label, and binding can be monitored by light microscopy, flow cytometry, fluorimetry, or other techniques known in the art. Such binding assays are performed essentially as described above.
  • In situ detection of antibody binding to the marker gene products can be performed, for example, by immunofluorescence or immunoelectron microscopy.
  • a histological specimen is removed from the patient, and a labeled antibody is applied to it, preferably by overlaying the antibody on a biological sample. This procedure also allows for determining the distribution of the marker gene product in the tissue examined. It will be apparent to those skilled in the art that a wide variety of histological methods are readily available for in situ detection.
  • Probes based on an expressed sequence tag (EST) identified from the Incyte Pharmaceuticals database due to homology with VEGF were used to screen a cDNA library derived from the human glioma cell line G61.
  • EST expressed sequence tag
  • Incyte Clone "INC1302516" was used to generate the following four probes:
  • VEGF-E-IgG expression plasmid was constructed by cloning the ORF in front of the human Fc (IgG) sequence.
  • the VEGF-E-IgG plasmid was co-transfected with Baculogold Baculovirus TM DNA (Pharmingen) using Lipofectin TM (GibcoBRL) into 10 5 Sf9 cells grown in Hink's TM TNM-FH medium (JRH Biosciences) supplemented with 10% fetal bovine serum. Cells were incubated for 5 days at 28°C. The supernatant was harvested and subsequently used for the first viral amplification by infecting Sf9 cells at an approximate multiplicity of infection (MOI) of 10.
  • MOI multiplicity of infection
  • Blots of human poly(A)+ RNA from multiple adult and fetal tissues and tumor cell lines were obtained from Clontech (Palo Alto, CA). Hybridization was carried out using 32 P-labeled probes containing the entire coding region and washed in 0.1 x SSC, 0.1% SDS at 63°C.
  • VEGF-E mRNA was detectable in fetal lung, kidney, brain, and liver and in adult heart, placenta, liver, skeletal muscle, kidney, and pancreas. VEGF-E mRNA was also found in A549 lung adenocarcinoma and HeLa cervical adenocarcinoma cell lines.
  • In situ hybridization is a powerful and versatile technique for the detection and localization of nucleic acid sequences within cell or tissue preparations. It may be useful, for example, to identify sites of gene expression, analyze the tissue distribution of transcription, identify and localize viral infection, follow changes in specific mRNA synthesis, and aid in chromosome mapping.
  • In situ hybridization was performed following an optimized version of the protocol by Lu and Gillett, Cell Vision 1: 169-176 (1994 ), using PCR-generated 33 P-labeled riboprobes. Briefly, formalin-fixed, paraffin-embedded human tissues were sectioned, deparaffinized, deproteinated in proteinase K (20 g/ml) for 15 minutes at 37°C, and further processed for in situ hybridization as described by Lu and Gillett, supra . A ( 33 -P)UTP-labeled antisense riboprobe was generated from a PCR product of 980 bp (using the oligonucleotide primers indicated below) and hybridized at 55°C overnight. The slides were dipped in KODAK NTB2 TM nuclear track emulsion and exposed for 4 weeks.
  • the tubes were incubated at 37°C for one hour. A total of 1.0 ⁇ l RQ1 DNase was added, followed by incubation at 37°C for 15 minutes. A total of 90 ⁇ l TE (10 mM Tris pH 7.6/1 mM EDTA pH 8.0) was added, and the mixture was pipetted onto DE81 paper. The remaining solution was loaded in a MICROCON-50 TM ultrafiltration unit, and spun using program 10 (6 minutes). The filtration unit was inverted over a second tube and spun using program 2 (3 minutes). After the final recovery spin, a total of 100 ⁇ l TE was added. Then 1 ⁇ l of the final product was pipetted on DE81 paper and counted in 6 ml of BIOFLUOR II TM .
  • the probe was run on a TBE/urea gel. A total of 1-3 ⁇ l of the probe or 5 ⁇ l of RNA Mrk III was added to 3 ⁇ l of loading buffer. After heating on a 95°C heat block for three minutes, the gel was immediately placed on ice. The wells of gel were flushed, and the sample was loaded and run at 180-250 volts for 45 minutes. The gel was wrapped in plastic wrap (SARAN TM brand) and exposed to XAR film with an intensifying screen in a -70°C freezer one hour to overnight.
  • SARAN TM brand plastic wrap
  • the slides were deparaffinized, placed in s.c. H 2 O, and rinsed twice in 2 x SSC at room temperature, for 5 minutes each time.
  • the sections were deproteinated in 20 ⁇ g/ml proteinase K (500 ⁇ l of 10 mg/ml in 250 ml RNAse-free RNAse buffer; 37°C, 15 minutes) for human embryo tissue, or 8 x proteinase K (100 ⁇ l in 250 ml RNAse buffer, 37°C, 30 minutes) for formalin tissues. Subsequent rinsing in 0.5 x SSC and dehydration were performed as described above.
  • the slides were laid out in a plastic box lined with Box buffer (4 x SSC, 50% formamide).
  • the filter paper was saturated.
  • the tissue was covered with 50 ⁇ l of hybridization buffer (3.75 g dextran sulfate + 6 ml s.c. H 2 O), vortexed, and heated in the microwave for 2 minutes with the cap loosened. After cooling on ice, 18.75 ml formamide, 3.75 ml 20 x SSC, and 9 ml s.c. H 2 O were added, and the tissue was vortexed well and incubated at 42°C for 1-4 hours.
  • hybridization buffer 3.75 g dextran sulfate + 6 ml s.c. H 2 O
  • VEGF-E For the lower human fetal limb, there was expression of VEGF-E in developing lower limb bones at the edge of the cartilagenous strom (i.e., around the outside edge), in developing tendons, in vascular smooth muscle, and in cells embracing developing skeletal muscle myocytes and myotubes. Expression was also observed at the epiphyseal growth plate. There was human fetal lymph node expression of VEGF-E in the marginal sinus of developing lymph nodes. There was human fetal thymus expression in the subcapsular region of the thymic cortex, possibly representing either the subcapsular epithelial cells or the proliferating, double-negative thymocytes that are found in this region. The human fetal spleen was negative for expression.
  • VEGF-E Trachea expression of VEGF-E in the smooth muscle of human fetal tissue was observed.
  • human fetal brain Cerebral cortex
  • the human fetal spinal cord was negative.
  • human fetal small intestine expression of VEGF-E in smooth muscle.
  • human fetal thyroid generalized expression of VEGF-E over thryoid epithelium The human fetal adrenal gland was negative.
  • Liver expression of VEGF-E in human fetal ductal plate cells was observed, as well as human fetal stomach expression in mural smooth muscle and human fetal skin expression in basal layer of the squamous epithelium.
  • human fetal placenta expression of VEGF-E in interstitial cells in trophoblastic villi was observed in the wall of the arteries and veins.
  • the expression pattern suggests that VEGF-E may be involved in cell differentiation and/or proliferation.
  • Expression patterns in developing skeletal muscle suggest that the protein may be involved in myoblast differentiation and/or proliferation.
  • Mouse embryonic fibroblast C3HlOT1/2 cells (ATCC) were grown in 50:50 Ham's F-12: low glucose DMEM medium containing 10% fetal calf serum (FCS). Cells were plated in duplicate in a 24-well plate at 1000, 2000, and 4000 cells/well. After 48 hours, cells were switched to medium containing 2% FCS and were incubated for 72 hours with 200, 800, or 2000 ng/ml VEGF-E or no growth factor added.
  • FCS fetal calf serum
  • Human umbilical vein endothelial cells (HUVEC, Cell Systems) were maintained in Complete Media (Cell Systems) and plated in triplicate in serum-free medium (Basic Media from Cell Systems containing 0.1% BSA) at 20,000 cells/well of a 48-well plate. Cells were incubated for 5 days with 200 or 400 ng/ml VEGF-E-IgG, 100 ng/ml VEGF, 20 ng/ml basic FGF, or no addition.
  • VEGF-E survival was 2-3 times greater with VEGF-E as compared to lack of growth factor addition.
  • VEGF and basic FGF were included as positive controls.
  • This assay follows the assay described in Davis and Camarillo, Experimental Cell Research, 224:39-51 (1996 ), or one modified from it as follows:
  • This assay will identify factors that facilitate cell survival in a 3-dimensional matrix in the presence of exogenous growth factors (VEGF, bFGF without PMA).
  • a positive result is equal to or less than 1.
  • 0 no apoptosis
  • 1 less than 20% cells are apoptotic
  • 2 less than 50% cells are apoptotic
  • 3 greater than 50% cells are apoptotic.
  • Stimulators of apoptosis in this system are expected to be apoptotic factors, and inhibitors are expected to prevent or lessen apoptosis.
  • a positive result is equal to or greater than 2.
  • 1 vacuoles present in less than 20% of cells
  • 2 vacuoles present in 20-50% of cells
  • 3 vacuoles present in greater than 50% of cells.
  • This assay is designed to identify factors that are involved in stimulating pinocytosis, ion pumping, permeability, and junction formation.
  • This assay is to identify factors that stimulate endothelial tube formation in a 3-dimensional matrix. This assay will identify factors that stimulate endothelial cells to differentiate into a tube-like structure in a 3-dimensional matrix in the presence of exogenous growth factors (VEGF, bFGF).
  • VEGF exogenous growth factors
  • a positive result is equal to or greater than 2.
  • Fig. 3A shows the HUVEC tube formation when no growth factors are present.
  • Fig. 3B shows where VEGF/bFGF, and PMA are present
  • Fig. 3C shows where VEGF and bFGF are present
  • Fig. 3D shows where VEGF and PMA are present
  • Fig. 3E shows where bFGF and PMA are present
  • Fig. 3F shows where VEGF is present
  • Fig. 3G shows where bFGF is present
  • Fig. 3H shows where PMA is present.
  • Figs. 4A and 4B show, respectively, the effect on HUVEC tube formation of VEGF-E-IgG at 1% dilution and of a buffer control (10 mM HEPES/0.14M NaCl/4% mannitol, pH 6.8) at 1% dilution.
  • Figs. 5A and 5B show, respectively, the effect on HUVEC tube formation of VEGF-E-poly-his at 1% dilution and of the buffer control used for VEGF-E-IgG at 1% dilution.
  • Transgenic mice were generated by microinjection of C57B1/6/SJL F2 mouse embryos (DNAX) with a vector suitable for such microinjection containing the cDNA encoding VEGF-E under the control of a keratin promoter ( Xie et al., Nature, 391: 90-92 (1998 )), driving expression in the skin.
  • DNAX C57B1/6/SJL F2 mouse embryos
  • mice Transgenic pups were wrinkled and shiny at birth and were delayed in getting their hair. The mice lost their phenotype by two weeks of age. There were no detectable histopathic changes.
  • Polyclonal antisera were generated in female New Zealand White rabbits against human VEGF-E.
  • the protein was homogenized with Freund's complete adjuvant for the primary injection and with Freund's incomplete adjuvant for all subsequent boosts.
  • 3.3 ⁇ g per kg body weight was injected directly into the popliteal lymph nodes, according to Bennett et al., J. Biol. Chem., 266: 23060-23067 (1991 ); and " Production of Antibodies by Inoculation into Lymph Nodes" by Sigel Sinha and VanderLaan in Methods in Enzymology, Vol. 93 (New York: Academic Press, 1983 ).
  • EXAMPLE 11 Inhibition of VEGF-stimulated endothelial cell (ACE cells) growth
  • Bovine adrenal cortical capillary endothelial cells (from primary culture, maximum of 12-14 passages) were plated in 96- well plates at 500 cells/well per 100 microliter. Assay media included low glucose DMEM, 10% calf serum, 2 mM glutamine, and 1X penicillin/streptomycin/fungizone.
  • Control wells included the following: (1) no ACE cells added; (2) ACE cells alone; (3) ACE cells plus 5 ng/ml FGF; (4) ACE cells plus 3 ng/ml VEGF; (5) ACE cells plus 3 ng/ml VEGF plus 1 ng/ml TGF-beta; and (6) ACE cells plus 3 ng/ml VEGF plus 5 ng/ml LIF.
  • the test sample poly-his tagged VEGF-E polypeptide (described in the Examples above; in 100 microliter volumes), was then added to the wells (at dilutions of 1%, 0.1% and 0.01%, respectively). The cell cultures were incubated for 6 - 7 days at 37°C/5% CO 2 .
  • the activity of VEGF-E was calculated as the percent inhibition of VEGF (3 ng/ml) stimulated proliferation (as determined by the acid phosphatase activity at OD 405 nm) relative to the cells without stimulation.
  • TGF-beta was employed as an activity reference- at 1 ng/ml, TGF-beta blocks 70-90% of VEGF-stimulated ACE cell proliferation. Results of the assay were interpreted as "positive" if the observed inhibition was ⁇ 30%.
  • VEGF-E In a first assay run, the VEGF-E at 1%, 0.1%, and 0.01% dilutions exhibited 52%, 90% and 96% inhibition, respectively. In a second assay run, the VEGF-E at 1%, 0.1%, and 0.01% dilutions exhibited 57%, 93% and 91% inhibition, respectively.

Claims (33)

  1. Acide nucléique isolé comprenant une séquence de nucléotides codant pour un polypeptide facteur de croissance de cellule endothéliale vasculaire E (VDGF-E), comprenant les résidus d'aminoacides 1 à 345 de la figure 2 (SEQ ID N° 2).
  2. Acide nucléique suivant la revendication 1, comprenant la séquence de nucléotides 259 à 1293 de la figure 1 (SEQ ID N° 1) ou son complément.
  3. Acide nucléique isolé comprenant une séquence de nucléotides qui s'hybride dans des conditions drastiques à la séquence de nucléotides de la revendication 1, les conditions drastiques comprenant une hybridation dans une solution à 50 % de formamide avec du SSC 5 x (NaCl 0,75 M, citrate de sodium 0,075 M), 50 mM de phosphate de sodium (pH 6/8), 0,1 % de pyrophosphate de sodium, de la solution de Denhardt 5 x, de l'ADN de sperme de saumon traité par ultrasons (50 µg/ml), 0,1 % de SDS et 10 % de sulfate de dextranne à 42°C, avec des lavages à 42°C dans une solution de SSC 0,2 x avec 0,1 % de SDS ; ou une hybridation en utilisant un tampon à 10 % de sulfate de dextranne, du SSC 2 x et 50 % de formamide à 55°C, avec ensuite un lavage dans des conditions hautement drastiques consistant en l'utilisation d'une solution de SSC 0,1 x contenant de l'EDTA à 55°C ;
    dans lequel ladite séquence de nucléotides code pour une protéine ayant une ou plusieurs des activités suivantes :
    (i) activation de la croissance sélective et/ou de la survie de cellules endothéliales de veine ombilicale humaine ;
    (ii) induction du gène précoce immédiat c-fos dans des lignées de cellules endothéliales humaines ;
    (iii) induction d'une hypertrophie des myocytes dans les cellules cardiaques ; ou
    (iv) inhibition de la prolifération, stimulée par le VEGF, des cellules endothéliales des capillaires du cortex surrénal.
  4. Acide nucléique isolé comprenant une séquence de nucléotides qui présente une identité de séquence d'acide nucléique de 70 % avec la séquence de la SEQ ID N° 1, positions 259 à 1293, l'identité de séquence étant déterminée sur la longueur totale des séquences comparées, et ladite séquence de nucléotides codant pour une protéine ayant une ou plusieurs des activités suivantes :
    (i) activation de la croissance sélective et/ou de la survie de cellules endothéliales de veine ombilicale humaine ;
    (ii) induction du gène précoce immédiat c-fos dans des lignées de cellules endothéliales humaines ;
    (iii) induction d'une hypertrophie des myocytes dans les cellules cardiaques ; ou
    (iv) inhibition de la prolifération, stimulée par le VEGF, des cellules endothéliales des capillaires du cortex surrénal.
  5. Vecteur comprenant l'acide nucléique de l'une quelconque des revendications 1 à 4.
  6. Cellule hôte comprenant l'acide nucléique de l'une quelconque des revendications 1 à 4.
  7. Cellule hôte suivant la revendication 6, ladite cellule étant une cellule d'ovaire de hamster chinois, une cellule d'insecte, une cellule de E. coli ou une cellule de levure.
  8. Cellule hôte suivant la revendication 7, qui est une cellule d'insecte infectée par un baculovirus.
  9. Procédé pour la production d'un polypeptide facteur de croissance de cellule endothéliale vasculaire E (VEGF-E), comprenant la culture de la cellule hôte de la revendication 6 dans des conditions convenables pour l'expression du polypeptide VEGF-E et la récupération du polypeptide VEGF-E à partir de la culture cellulaire.
  10. Polypeptide VEGF-E codé par l'acide nucléique de l'une quelconque des revendications 1 à 4, pouvant être produit par le procédé de la revendication 9.
  11. Polypeptide VEGF-E comprenant les résidus d'aminoacides 1 à 345 de la figure 2 (SEQ ID N° 2).
  12. Polypeptide VEGF-E choisi dans le groupe consistant en :
    (a) un polypeptide comprenant les résidus d'aminoacides 1 à 345 de la figure 2 (SEQ ID N° 2) ; et
    (b) un fragment polypeptidique de (a), ledit fragment ayant une ou plusieurs des activités suivantes :
    (i) activation de la croissance sélective et/ou de la survie de cellules endothéliales de veine ombilicale humaine ;
    (ii) induction du gène précoce immédiat c-fos dans des lignées de cellules endothéliales humaines ;
    (iii) induction d'une hypertrophie des myocytes dans les cellules cardiaques ; ou
    (iv) inhibition de la prolifération, stimulée par le VEGF, des cellules endothéliales des capillaires du cortex surrénal.
  13. Polypeptide VEGF-E codé par le segment d'insertion de séquence de nucléotides ayant le numéro de dépôt ATCC 209653.
  14. Polypeptide chimère comprenant le polypeptide de l'une quelconque des revendications 10 à 13 fusionné à une séquence d'aminoacides hétérologue.
  15. Polypeptide chimère suivant la revendication 14, dans lequel ladite séquence d'aminoacides hétérologue est une séquence de marqueur épitopique ou une région Fc d'une immunoglobuline.
  16. Composition comprenant le polypeptide de l'une quelconque des revendications 10 à 15 en mélange avec un support.
  17. Composition suivant la revendication 16, comprenant une quantité thérapeutiquement efficace du polypeptide, dans laquelle le support est un support pharmaceutiquement acceptable.
  18. Polypeptide suivant l'une quelconque des revendications 10 à 15, destiné à être utilisé dans une méthode de traitement.
  19. Polypeptide suivant la revendication 18, destiné à être utilisé dans une méthode de traitement, le traitement étant le traitement de plaies, d'un trouble en rapport avec le tissu osseux, ou dans la formation ou la régénération du tissu musculaire.
  20. Utilisation d'un polypeptide VEGF-E tel que défini dans l'une quelconque des revendications 10 à 15, dans la production d'un médicament destiné à être utilisé dans le traitement de plaies, dans le traitement d'un trouble en rapport avec le tissu osseux, ou dans la formation ou régénération du tissu musculaire.
  21. Produit pharmaceutique comprenant :
    (a) la composition de la revendication 16 ;
    (b) un récipient contenant ledit polypeptide ; et
    (c) une étiquette fixée audit récipient, ou un encart d'emballage incorporé audit produit pharmaceutique, mentionnant l'utilisation dudit polypeptide VEGF-E dans le traitement d'un trouble cardio-vasculaire ou endothélial.
  22. Méthode pour le diagnostic de troubles cardio-vasculaires et endothéliaux chez un mammifère, comprenant la détection du degré d'expression d'un gène codant pour un polypeptide facteur de croissance de cellules endothéliales vasculaires E (VEGF-E) tel que défini dans l'une quelconque des revendications 10 à 13 (a) dans un échantillon d'essai de cellules d'un tissu obtenues à partir du mammifère, et (b) dans un échantillon témoin de cellules de tissu normal connu du même type cellulaire, dans laquelle un plus haut ou plus bas degré d'expression dans l'échantillon d'essai indique la présence d'un dysfonctionnement cardio-vasculaire ou endothélial chez le mammifère à partir duquel les cellules de tissu d'essai ont été obtenues.
  23. Composé qui inhibe l'expression ou l'activité d'un polypeptide VEGF-E tel que défini dans l'une quelconque des revendications 10 à 13, composé qui est un anticorps qui se lie spécifiquement audit polypeptide VEGF-E, un acide nucléique antisens, un acide nucléique en triple hélice ou un ribozyme.
  24. Anticorps isolé qui se lie à un polypeptide facteur de croissance de cellules endothéliales vasculaires E (VEGF-E) tel que défini dans l'une quelconque des revendications 10 à 13.
  25. Anticorps suivant la revendication 24, qui est un anticorps monoclonal.
  26. Méthode pour déterminer la présence d'un polypeptide facteur de croissance de cellules endothéliales vasculaires E (VEGF-E) tel que défini dans l'une quelconque des revendications 10 à 13, comprenant l'exposition d'une cellule supposée contenir le polypeptide VEGF-E à l'anticorps de la revendication 24 et la détermination de la liaison dudit anticorps à ladite cellule.
  27. Méthode pour le diagnostic de troubles cardio-vasculaires, endothéliaux ou angiogéniques chez un mammifère, comprenant (a) la mise en contact de l'anticorps de la revendication 24 ou de la revendication 25 avec un échantillon d'essai de cellules d'un tissu obtenues à partir du mammifère, et (b) la détection de la formation d'un complexe entre l'anticorps anti VEGF-E et le polypeptide VEGF-E dans l'échantillon d'essai.
  28. Composé suivant la revendication 23 ou anticorps suivant la revendication 24 ou la revendication 25, destiné à être utilisé dans une méthode de traitement.
  29. Composé ou anticorps suivant la revendication 28, destiné à être utilisé dans une méthode de traitement, le traitement étant l'inhibition de l'angiogenèse.
  30. Composé ou anticorps suivant la revendication 29, destiné à être utilisé dans une méthode de traitement, le traitement étant le traitement d'une tumeur ou d'un trouble de la rétine.
  31. Utilisation du composé de la revendication 23 ou de l'anticorps de la revendication 24 ou de la revendication 25, dans la production d'un médicament destiné à être utilisé dans le traitement de l'angiogenèse.
  32. Utilisation suivant la revendication 31, dans laquelle le traitement est le traitement d'une tumeur ou d'un trouble de la rétine.
  33. Article manufacturé, comprenant :
    (a) un récipient ;
    (b) une étiquette sur le récipient ; et
    (c) une composition comprenant l'anticorps de la revendication 24 ou de la revendication 25 présente dans le récipient ; dans lequel l'étiquette sur le récipient fournit des instructions pour l'utilisation de l'anticorps dans une méthode thérapeutique ou une méthode de diagnostic.
EP99912362A 1998-03-17 1999-03-10 Polypeptides homologues aux vegf et bmp1 Expired - Lifetime EP1064382B1 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
DK08012659.2T DK2016951T3 (da) 1998-03-17 1999-03-10 VEGF- og BMP1-homologe polypeptider
EP08012659A EP2016951B1 (fr) 1998-03-17 1999-03-10 Polypeptides homologues à VEGF et BMP1
CY20081101286T CY1108500T1 (el) 1998-03-17 2008-11-11 Πολυπεπτιδια ομολογα με vefg και bmp1

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US40220 1998-03-17
US09/040,220 US6391311B1 (en) 1998-03-17 1998-03-17 Polypeptides having homology to vascular endothelial cell growth factor and bone morphogenetic protein 1
US18421698A 1998-11-02 1998-11-02
US184216 1998-11-02
PCT/US1999/005190 WO1999047677A2 (fr) 1998-03-17 1999-03-10 Polypeptides homologues de vegf et bmp1

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP08012659A Division EP2016951B1 (fr) 1998-03-17 1999-03-10 Polypeptides homologues à VEGF et BMP1

Publications (2)

Publication Number Publication Date
EP1064382A2 EP1064382A2 (fr) 2001-01-03
EP1064382B1 true EP1064382B1 (fr) 2008-08-20

Family

ID=26716852

Family Applications (2)

Application Number Title Priority Date Filing Date
EP08012659A Expired - Lifetime EP2016951B1 (fr) 1998-03-17 1999-03-10 Polypeptides homologues à VEGF et BMP1
EP99912362A Expired - Lifetime EP1064382B1 (fr) 1998-03-17 1999-03-10 Polypeptides homologues aux vegf et bmp1

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP08012659A Expired - Lifetime EP2016951B1 (fr) 1998-03-17 1999-03-10 Polypeptides homologues à VEGF et BMP1

Country Status (14)

Country Link
US (11) US6455283B1 (fr)
EP (2) EP2016951B1 (fr)
JP (3) JP4532733B2 (fr)
AT (1) ATE405651T1 (fr)
AU (1) AU758353B2 (fr)
CA (1) CA2322792C (fr)
CY (2) CY1108500T1 (fr)
DE (1) DE69939374D1 (fr)
DK (2) DK1064382T3 (fr)
ES (2) ES2313778T3 (fr)
HK (1) HK1125853A1 (fr)
IL (5) IL138488A0 (fr)
PT (1) PT2016951E (fr)
WO (1) WO1999047677A2 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA018848B1 (ru) * 2010-10-20 2013-11-29 Вадим Юльевич ШАНИН Иммунологически активное вещество, способ его получения, биозонд на его основе и способ диагностики состояния здоровья с применением биозонда, фармацевтическая композиция и способ лечения с использованием фармацевтической композиции
WO2014159813A1 (fr) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Molécules polynucléotidiques à longue durée de vie

Families Citing this family (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000053758A2 (fr) * 1999-03-08 2000-09-14 Genentech, Inc. Compositions et methodes de traitement des maladies immunitaires
WO2000053760A2 (fr) * 1999-03-12 2000-09-14 Genentech, Inc. Technique permettant de prevenir la mort des neurones retiniens et traitement des maladies oculaires
AU2678999A (en) 1998-02-13 1999-08-30 Genetrace Systems, Inc. Use of ribozymes for functionating genes
JP2002505144A (ja) * 1998-03-06 2002-02-19 クロスカート インコーポレイテッド 軟組織異種移植片
ES2313778T3 (es) 1998-03-17 2009-03-01 Genentech, Inc. Polipeptidos homologos de vegf y de bmp1.
EP0984063A3 (fr) * 1998-08-31 2000-08-16 Eli Lilly And Company Gene et proteine apparentee au vegf
EP1988102B1 (fr) * 1998-12-07 2013-08-21 ZymoGenetics, Inc. Facteur de croissance homologue ZVEGF3
US6432673B1 (en) 1998-12-07 2002-08-13 Zymogenetics, Inc. Growth factor homolog ZVEGF3
US6663870B2 (en) 1998-12-07 2003-12-16 Zymogenetics, Inc. Methods for promoting growth of bone using zvegf3
US6783953B1 (en) 1998-12-22 2004-08-31 Janssen Pharmaceutica N.V. Vascular endothelial growth factor-X
DE69936733T2 (de) * 1998-12-22 2008-06-05 Janssen Pharmaceutica N.V. Vaskulärer endothelialer wachstumsfaktor x
EP1710299A3 (fr) * 1998-12-30 2007-01-10 Millennium Pharmaceuticals, Inc. Proteines secretées et polynucleotides codant pour ces proteines
AU4008500A (en) * 1999-04-06 2000-10-23 Eli Lilly And Company Platelet-derived growth factor related gene and protein
US6630142B2 (en) 1999-05-03 2003-10-07 Zymogenetics, Inc. Method of treating fibroproliferative disorders
AU7868000A (en) * 1999-10-07 2001-05-10 Curagen Corporation Growth factor polypeptides and nucleic acids encoding same
US6893637B1 (en) * 1999-10-21 2005-05-17 Zymogenetics, Inc. Method of treating fibrosis
WO2001028586A1 (fr) * 1999-10-21 2001-04-26 Zymogenetics, Inc. Traitement de la fibrose
AU1430601A (en) * 1999-11-02 2001-05-14 Eli Lilly And Company Methods of using lp8, a pdgf-related protein, to treat musculoskeletal disorders
CA2397919A1 (fr) * 2000-01-19 2001-07-26 Parkash S. Gill Compositions renfermant des oligonucleotides antisens diriges contre le vegf et methodes associees
US7141607B1 (en) 2000-03-10 2006-11-28 Insite Vision Incorporated Methods and compositions for treating and inhibiting retinal neovascularization
WO2001072132A1 (fr) * 2000-03-28 2001-10-04 Ludwig Institute For Cancer Research Animaux transgeniques non humains exprimant un facteur de croissance c derive de plaquettes (pdgf-c) et utilisations
GB0008269D0 (en) * 2000-04-05 2000-05-24 Astrazeneca Ab Combination chemotherapy
US6759386B2 (en) * 2000-04-06 2004-07-06 Wayne P. Franco Methods of use of fibroblast growth factor, vascular endothelial growth factor and related proteins in the treatment of acute and chronic heart disease
US20070111935A1 (en) * 2000-04-06 2007-05-17 Franco Wayne P Combination growth factor therapy and cell therapy for treatment of acute and chronic diseases of the organs
US20050277576A1 (en) * 2000-04-06 2005-12-15 Franco Wayne P Combination growth factor therapy and cell therapy for treatment of acute and chronic diseases of the organs
US7166280B2 (en) * 2000-04-06 2007-01-23 Franco Wayne P Combination growth factor therapy and cell therapy for treatment of acute and chronic heart disease
PT2277528E (pt) * 2000-04-12 2015-02-05 Life Sciences Res Partners Vzw Uso de vegf e homólogos para tratar doenças do neurónio
AU2001265953B2 (en) * 2000-05-12 2005-09-29 D. Collen Research Foundation Vzw Use of inhibitors of placental growth factor for the treatment of pathological angiogenesis, pathological arteriogenesis, inflammation, tumour formation and/or vascular leakage
AU2001276934A1 (en) * 2000-07-18 2002-02-05 Joslin Diabetes Center Inc. Methods of modulating fibrosis
US20020091082A1 (en) * 2000-09-13 2002-07-11 Aiello Lloyd P. Methods of modulating symptoms of hypertension
US20040086507A1 (en) * 2000-10-19 2004-05-06 Kenya Shitara Antibody inhibiting vplf activity
AU2884102A (en) 2000-12-07 2002-06-18 Sangamo Biosciences Inc Regulation of angiogenesis with zinc finger proteins
US7067317B2 (en) 2000-12-07 2006-06-27 Sangamo Biosciences, Inc. Regulation of angiogenesis with zinc finger proteins
US20020141970A1 (en) * 2001-03-05 2002-10-03 Pettit Dean K. Stable aqueous solutions of granulocyte macrophage colony-stimulating factor
KR20080083072A (ko) 2001-03-09 2008-09-12 얀센 파마슈티카 엔.브이. Vegf-x 또는 그의 길항제에 의한 평활근 세포 증식의 조절
US6913762B2 (en) * 2001-04-25 2005-07-05 Mayo Foundation For Medical Education And Research Stent having non-woven framework containing cells
US7981863B2 (en) 2001-09-19 2011-07-19 Neuronova Ab Treatment of Parkinson's disease with PDGF
CN1602207A (zh) * 2001-12-11 2005-03-30 法布罗根股份有限公司 抑制眼病理过程的方法
AU2003217531A1 (en) * 2002-05-02 2003-11-17 Massachusetts Eye And Ear Infirmary Ocular drug delivery systems and use thereof
JP2005524710A (ja) * 2002-05-06 2005-08-18 ジェネンテック・インコーポレーテッド 骨欠陥の治療へのvegfの用途
AU2003245935A1 (en) * 2002-06-13 2003-12-31 Novuspharma S.P.A. Derivatives of chromen-2-one as inhibitors of vegf production in mammalian cells
US20040052161A1 (en) * 2002-09-17 2004-03-18 Steven Liao Mechanical clock having wireless manipulation and adjustment function
AU2003288027A1 (en) 2002-11-16 2004-06-15 Dade Behring Marburg Gmbh Scd40l, papp-a and placental growth factor (plgf) used as a biochemical marker combination in cardiovascular diseases
WO2005069906A2 (fr) * 2004-01-16 2005-08-04 Yale University Procedes et compositions pour le traitement de maladies inflammatoires liees au facteur de croissance endothelial vasculaire et mediees par les th2
WO2005087177A2 (fr) * 2004-03-05 2005-09-22 Ludwig Institute For Cancer Research Anticorps chimeriques anti-vegf-d et anticorps humanises anti-vegf-d et leurs procedes d'utilisation
WO2005087812A1 (fr) * 2004-03-05 2005-09-22 Ludwig Institute For Cancer Research Substances d'anticorps polyvalentes et procedes pour la famille vegf/pdgf des facteurs de croissance
US8415315B2 (en) 2004-05-06 2013-04-09 University Of Central Florida Research Foundation, Inc. Methods and compositions for inhibiting the proliferation of cancer cells
RS51481B (en) * 2004-05-27 2011-04-30 Vib, Vzw TREATMENT OF AMIOTROPHIC LATERAL SCLEROSIS
US20060045902A1 (en) * 2004-09-01 2006-03-02 Serbousek Jon C Polymeric wrap for in vivo delivery of osteoinductive formulations
DE102004051847B4 (de) * 2004-10-25 2008-09-18 Dade Behring Marburg Gmbh Verhältnis von PIGF und Flt-1 als prognostischer Parameter bei kardio-vaskulären Erkrankungen
ZA200707258B (en) * 2005-03-24 2008-06-25 Thromb X N V Novel anti-PLGF antibody
ES2363758T3 (es) * 2005-08-15 2011-08-16 Vegenics Pty Ltd Vegf y pdgf modificados con propiedades angiogénicas mejoradas.
US8114399B2 (en) 2006-05-17 2012-02-14 Ludwig Institute For Cancer Research Targeting VEGF-B regulation of fatty acid transporters to modulate human diseases
ES2354922B1 (es) 2009-09-02 2012-02-07 Fundacion Institut De Recerca De L'hospital Universitari Vall D'hebron Marcadores para la selección de terapias personalizadas para el tratamiento del c�?ncer.
TW201302793A (zh) 2010-09-03 2013-01-16 Glaxo Group Ltd 新穎之抗原結合蛋白
ES2628321T3 (es) 2011-12-01 2017-08-02 Thrombogenics N.V. Mejora del resultado de una trabeculectomía
NZ724395A (en) 2012-04-24 2018-03-23 Thrombogenics Nv Anti-pdgf-c antibodies
WO2013172721A1 (fr) * 2012-05-18 2013-11-21 Wise Lyn Marie Traitements combinés et compositions pour la cicatrisation de plaies
CN105713095B (zh) * 2016-03-14 2021-05-07 南京安吉生物科技有限公司 一种多功能融合多肽及其制备方法和应用
KR101943081B1 (ko) 2017-08-31 2019-01-29 (주)케어젠 주름 개선 활성을 나타내는 펩타이드 및 이의 용도

Family Cites Families (122)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2035733A (en) * 1935-06-10 1936-03-31 Marathon Electric Mfg Fan motor mounting
US2813673A (en) * 1953-07-09 1957-11-19 Gilbert Co A C Tiltable oscillating fan
US2765977A (en) * 1954-10-13 1956-10-09 Morrison Hackley Electric ventilating fans
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4263428A (en) 1978-03-24 1981-04-21 The Regents Of The University Of California Bis-anthracycline nucleic acid function inhibitors and improved method for administering the same
JPS6023084B2 (ja) 1979-07-11 1985-06-05 味の素株式会社 代用血液
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
ZA811368B (en) 1980-03-24 1982-04-28 Genentech Inc Bacterial polypedtide expression employing tryptophan promoter-operator
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
DE3169595D1 (en) 1980-11-10 1985-05-02 Gersonde Klaus Method of preparing lipid vesicles by ultrasonic treatment, the use of this method and apparatus for its application
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
NZ201705A (en) 1981-08-31 1986-03-14 Genentech Inc Recombinant dna method for production of hepatitis b surface antigen in yeast
US4568243A (en) * 1981-10-08 1986-02-04 Barry Wright Corporation Vibration isolating seal for mounting fans and blowers
US4640835A (en) 1981-10-30 1987-02-03 Nippon Chemiphar Company, Ltd. Plasminogen activator derivatives
JPS58118008A (ja) 1982-01-06 1983-07-13 Nec Corp デ−タ処理装置
EP0088046B1 (fr) 1982-02-17 1987-12-09 Ciba-Geigy Ag Lipides en phase aqueuse
DE3218121A1 (de) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Arzneimittel zur tumorbehandlung
EP0102324A3 (fr) 1982-07-29 1984-11-07 Ciba-Geigy Ag Lipides et composés tensio-actifs en phase aqueuse
US4870009A (en) 1982-11-22 1989-09-26 The Salk Institute For Biological Studies Method of obtaining gene product through the generation of transgenic animals
AU2353384A (en) 1983-01-19 1984-07-26 Genentech Inc. Amplification in eukaryotic host cells
US4713339A (en) 1983-01-19 1987-12-15 Genentech, Inc. Polycistronic expression vector construction
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4675187A (en) 1983-05-16 1987-06-23 Bristol-Myers Company BBM-1675, a new antibiotic complex
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
HUT35524A (en) 1983-08-02 1985-07-29 Hoechst Ag Process for preparing pharmaceutical compositions containing regulatory /regulative/ peptides providing for the retarded release of the active substance
ATE59966T1 (de) 1983-09-26 1991-02-15 Ehrenfeld Udo Mittel und erzeugnis fuer die diagnose und therapie von tumoren sowie zur behandlung von schwaechen der zelligen und humoralen immunabwehr.
US4615885A (en) 1983-11-01 1986-10-07 Terumo Kabushiki Kaisha Pharmaceutical composition containing urokinase
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4736866B1 (en) 1984-06-22 1988-04-12 Transgenic non-human mammals
EP0206448B1 (fr) 1985-06-19 1990-11-14 Ajinomoto Co., Inc. Hémoglobine liée à un poly(oxyde d'alkylène)
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4703152A (en) * 1985-12-11 1987-10-27 Holmes Products Corp. Tiltable and adjustably oscillatable portable electric heater/fan
EP0272253A4 (fr) 1986-03-07 1990-02-05 Massachusetts Inst Technology Procede pour ameliorer la stabilite des glycoproteines.
US4791192A (en) 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
US5013649A (en) 1986-07-01 1991-05-07 Genetics Institute, Inc. DNA sequences encoding osteoinductive products
IL83003A (en) * 1986-07-01 1995-07-31 Genetics Inst Factors that soak bone formation
US5631142A (en) 1986-07-01 1997-05-20 Genetics Institute, Inc. Compositions comprising bone morphogenetic protein-2 (BMP-2)
US5543394A (en) 1986-07-01 1996-08-06 Genetics Institute, Inc. Bone morphogenetic protein 5(BMP-5) compositions
US4877864A (en) 1987-03-26 1989-10-31 Genetics Institute, Inc. Osteoinductive factors
US5106748A (en) 1986-07-01 1992-04-21 Genetics Institute, Inc. Dna sequences encoding 5 proteins
US4850804A (en) * 1986-07-07 1989-07-25 Tatung Company Of America, Inc. Portable electric fan having a universally adjustable mounting
ATE76311T1 (de) 1986-08-19 1992-06-15 Genentech Inc Einrichtung und dispersion zum intrapulmonalen eingeben von polypeptidwuchsstoffen und zytokinen.
EP0266032A1 (fr) 1986-08-29 1988-05-04 Beecham Group Plc Enzyme fibrinolytique modifiée
US5010182A (en) 1987-07-28 1991-04-23 Chiron Corporation DNA constructs containing a Kluyveromyces alpha factor leader sequence for directing secretion of heterologous polypeptides
ES2058199T3 (es) 1987-09-23 1994-11-01 Bristol Myers Squibb Co Heteroconjugados de anticuerpos para la eliminacion de celulas infectadas por el vih.
GB8724885D0 (en) 1987-10-23 1987-11-25 Binns M M Fowlpox virus promotors
US5283187A (en) 1987-11-17 1994-02-01 Brown University Research Foundation Cell culture-containing tubular capsule produced by co-extrusion
US4892538A (en) 1987-11-17 1990-01-09 Brown University Research Foundation In vivo delivery of neurotransmitters by implanted, encapsulated cells
WO1989006692A1 (fr) 1988-01-12 1989-07-27 Genentech, Inc. Procede de traitement de cellules tumorales par inhibition de la fonction receptrice du facteur de croissance
AU4005289A (en) 1988-08-25 1990-03-01 Smithkline Beecham Corporation Recombinant saccharomyces
US5240848A (en) 1988-11-21 1993-08-31 Monsanto Company Dna sequences encoding human vascular permeability factor having 189 amino acids
US5147638A (en) 1988-12-30 1992-09-15 Oklahoma Medical Research Foundation Inhibition of tumor growth by blockade of the protein C system
FR2646437B1 (fr) 1989-04-28 1991-08-30 Transgene Sa Nouvelles sequences d'adn, leur application en tant que sequence codant pour un peptide signal pour la secretion de proteines matures par des levures recombinantes, cassettes d'expression, levures transformees et procede de preparation de proteines correspondant
US5332671A (en) 1989-05-12 1994-07-26 Genetech, Inc. Production of vascular endothelial cell growth factor and DNA encoding same
CA2062795A1 (fr) 1989-06-29 1990-12-30 Michael W. Fanger Reactifs bispecifiques pour le traitement du sida
CA2020729A1 (fr) 1989-07-19 1991-01-20 Michael C. Kiefer Proteine morphogenetique des os
JP3045398B2 (ja) 1989-09-06 2000-05-29 武田薬品工業株式会社 蛋白質、dnaおよびその用途
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5126323A (en) 1989-11-16 1992-06-30 Genetics Institute, Inc. Homogeneous purified k-fgf and compositions containing the same
USD325435S (en) * 1990-09-24 1992-04-14 Vornado Air Circulation Systems, Inc. Fan support base
US5206161A (en) 1991-02-01 1993-04-27 Genentech, Inc. Human plasma carboxypeptidase B
JPH06507398A (ja) 1991-05-14 1994-08-25 リプリジェン コーポレーション Hiv感染治療のための異種複合抗体
US5382569A (en) 1991-05-16 1995-01-17 Warner-Lambert Company Endotherlin antagonists
EP0590058B1 (fr) 1991-06-14 2003-11-26 Genentech, Inc. ANTICORP HUMANISE specifique pour heregulin
ATE175441T1 (de) 1991-06-25 1999-01-15 Genetics Inst Bmp-9 zusammensetzungen
WO1993008829A1 (fr) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions induisant la destruction de cellules infectees par l'hiv
US5296769A (en) * 1992-01-24 1994-03-22 Electrolux Corporation Air guide assembly for an electric motor and methods of making
IL105914A0 (en) 1992-06-04 1993-10-20 Univ California Methods and compositions for in vivo gene therapy
GB9214857D0 (en) 1992-07-13 1992-08-26 Medical Res Council Human nucleic acid fragments and their use
EP1167384B1 (fr) * 1992-10-28 2006-12-13 Genentech, Inc. Récepteur du hVEGF comme antagoniste du VEGF
DK0669836T3 (da) 1992-11-13 1996-10-14 Idec Pharma Corp Terapeutisk anvendelse af kimære og radioaktivt mærkede antistoffer og humant B-lymfocytbegrænset differentieringsantigen til behandling af B-cellelymfom
US5310313A (en) * 1992-11-23 1994-05-10 Chen C H Swinging type of electric fan
ES2141128T3 (es) 1993-03-24 2000-03-16 Berlex Biosciences Combinacion de agentes anti-hormonales y moleculas de fijacion.
ATE312198T1 (de) 1993-04-05 2005-12-15 Univ Utah Res Found Diagnose und behandlung von supravalvular aortic stenosis
US5637480A (en) 1993-05-12 1997-06-10 Genetics Institute, Inc. DNA molecules encoding bone morphogenetic protein-10
US5641756A (en) 1993-07-27 1997-06-24 Hybridon, Inc. Modified VEGF oligonucleotides
JPH09501932A (ja) 1993-08-26 1997-02-25 ジェネティックス・インスティテュート・インコーポレイテッド ヒト・骨形態形成蛋白を用いる神経再生
US5773223A (en) 1993-09-02 1998-06-30 Chiron Corporation Endothelin B1, (ETB1) receptor polypeptide and its encoding nucleic acid methods, and uses thereof
ATE319823T1 (de) 1993-12-07 2006-03-15 Inst Genetics Llc Bmp-12, bmp-13 und diese enthaltende sehne- induzierende zusammensetzungen
US5399677A (en) 1993-12-07 1995-03-21 Genetics Institute, Inc. Mutants of bone morphogenetic proteins
DE69535221T2 (de) 1994-04-25 2007-09-13 Genentech, Inc., South San Francisco Cardiotrophin und verwendung davon
WO2000053758A2 (fr) 1999-03-08 2000-09-14 Genentech, Inc. Compositions et methodes de traitement des maladies immunitaires
US5454419A (en) 1994-09-19 1995-10-03 Polybore, Inc. Method for lining a casing
US5681746A (en) 1994-12-30 1997-10-28 Chiron Viagene, Inc. Retroviral delivery of full length factor VIII
IL117645A (en) 1995-03-30 2005-08-31 Genentech Inc Vascular endothelial cell growth factor antagonists for use as medicaments in the treatment of age-related macular degeneration
US5518370A (en) * 1995-04-03 1996-05-21 Duracraft Corporation Portable electric fan with swivel mount
SG68529A1 (en) 1995-05-25 1999-11-16 Univ Singapore The use of des-aspartate-angiotensin i (agr-val-tyr-ile-his-pro-phe-his-leu) as an anti-cardiac hypertrophic agent
US6365393B1 (en) * 1996-02-28 2002-04-02 Bayer Aktiengesellschaft Parapoxviruses containing foreign DNA, their production and their use in vaccines
AU2582897A (en) 1996-03-15 1997-10-01 Millennium Pharmaceuticals, Inc. Compositions and methods for the diagnosis, prevention, and treatment of neoplastic cell growth and proliferation
DE69740038D1 (fr) * 1996-07-12 2010-12-16 Genentech Inc
WO1998006724A1 (fr) 1996-08-09 1998-02-19 Yamanouchi Pharmaceutical Co., Ltd. Agonistes du recepteur du glutamate metabotropique
DK1749836T3 (da) 1996-08-23 2009-08-31 Vegenics Ltd Rekombinant vaskulær endothelcellevækstfaktor D
EP3260468A1 (fr) 1997-04-07 2017-12-27 Genentech, Inc. Anticorps anti-vegf
USD398983S (en) * 1997-08-08 1998-09-29 Vornado Air Circulation Systems, Inc. Fan
US6391311B1 (en) 1998-03-17 2002-05-21 Genentech, Inc. Polypeptides having homology to vascular endothelial cell growth factor and bone morphogenetic protein 1
WO1999037671A1 (fr) 1998-01-27 1999-07-29 Eli Lilly And Company Gene et proteine associes au facteur de croissance des cellules endotheliales vasculaires
PT1490386E (pt) 1998-03-10 2008-11-24 Genentech Inc Novos polipéptidos e ácidos nucleicos que os codificam
ES2313778T3 (es) 1998-03-17 2009-03-01 Genentech, Inc. Polipeptidos homologos de vegf y de bmp1.
WO2000004183A1 (fr) 1998-07-15 2000-01-27 Human Genome Sciences, Inc. Proteine morphogenetique osseuse
CN1330664A (zh) 1998-09-30 2002-01-09 路德维格癌症研究所 血小板衍生生长因子c、其编码dna及其应用
CA2347655A1 (fr) 1998-10-28 2000-05-04 Incyte Pharmaceuticals, Inc. Molecules apparentees au facteur de croissance
US20030027998A1 (en) 1998-10-30 2003-02-06 Holtzman Douglas A. Novel genes encoding proteins having prognostic, diagnostic, preventive, therapeutic, and other uses
JP3695642B2 (ja) 1998-12-01 2005-09-14 ジェネンテック・インコーポレーテッド 血管形成及び心臓血管新生の促進又は阻害
EP1988102B1 (fr) 1998-12-07 2013-08-21 ZymoGenetics, Inc. Facteur de croissance homologue ZVEGF3
US6432673B1 (en) * 1998-12-07 2002-08-13 Zymogenetics, Inc. Growth factor homolog ZVEGF3
DE69936733T2 (de) 1998-12-22 2008-06-05 Janssen Pharmaceutica N.V. Vaskulärer endothelialer wachstumsfaktor x
EP1710299A3 (fr) 1998-12-30 2007-01-10 Millennium Pharmaceuticals, Inc. Proteines secretées et polynucleotides codant pour ces proteines
US6155782A (en) * 1999-02-01 2000-12-05 Hsu; Chin-Tien Portable fan
AU4008500A (en) 1999-04-06 2000-10-23 Eli Lilly And Company Platelet-derived growth factor related gene and protein
AU4714500A (en) 1999-05-03 2000-11-17 Zymogenetics Inc. Growth factor homolog zvegf4
WO2001040714A1 (fr) * 1999-12-06 2001-06-07 The Holmes Group, Inc. Element chauffant pivotant
WO2003068155A2 (fr) * 2002-02-12 2003-08-21 Vanderbilt University Inhibition de la signalisation du recepteur vegf inversant la resistance tumorale a la radiotherapie
BR0201397B1 (pt) * 2002-04-19 2011-10-18 arranjo de montagem para um ventilador de refrigerador.
US6830433B2 (en) * 2002-08-05 2004-12-14 Kaz, Inc. Tower fan
US7059826B2 (en) * 2003-07-25 2006-06-13 Lasko Holdings, Inc. Multi-directional air circulating fan
US20050128698A1 (en) * 2003-12-10 2005-06-16 Huang Cheng Y. Cooling fan
WO2005087812A1 (fr) * 2004-03-05 2005-09-22 Ludwig Institute For Cancer Research Substances d'anticorps polyvalentes et procedes pour la famille vegf/pdgf des facteurs de croissance
US7088913B1 (en) * 2004-06-28 2006-08-08 Jcs/Thg, Llc Baseboard/upright heater assembly
CN2835669Y (zh) * 2005-09-16 2006-11-08 霍树添 立柱式电风扇的送风机构

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA018848B1 (ru) * 2010-10-20 2013-11-29 Вадим Юльевич ШАНИН Иммунологически активное вещество, способ его получения, биозонд на его основе и способ диагностики состояния здоровья с применением биозонда, фармацевтическая композиция и способ лечения с использованием фармацевтической композиции
WO2014159813A1 (fr) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Molécules polynucléotidiques à longue durée de vie

Also Published As

Publication number Publication date
JP2013224300A (ja) 2013-10-31
ES2389387T3 (es) 2012-10-25
DK1064382T3 (da) 2008-12-08
IL138488A0 (en) 2001-10-31
EP2016951A1 (fr) 2009-01-21
JP5473441B2 (ja) 2014-04-16
JP2010001292A (ja) 2010-01-07
CY1113126T1 (el) 2016-04-13
CY1108500T1 (el) 2014-04-09
US7371377B2 (en) 2008-05-13
US20040219640A1 (en) 2004-11-04
ATE405651T1 (de) 2008-09-15
CA2322792A1 (fr) 1999-09-23
US7494977B2 (en) 2009-02-24
HK1125853A1 (en) 2009-08-21
WO1999047677A3 (fr) 1999-11-11
IL189572A (en) 2013-01-31
AU758353B2 (en) 2003-03-20
WO1999047677A2 (fr) 1999-09-23
US7575879B2 (en) 2009-08-18
JP2004515201A (ja) 2004-05-27
US20100331251A1 (en) 2010-12-30
EP2016951B1 (fr) 2012-06-27
US20090264370A9 (en) 2009-10-22
US20080299119A1 (en) 2008-12-04
US20090012003A1 (en) 2009-01-08
IL189573A0 (en) 2009-02-11
US20040235740A1 (en) 2004-11-25
US20070031929A1 (en) 2007-02-08
IL189572A0 (en) 2009-02-11
PT2016951E (pt) 2012-09-27
IL189571A0 (en) 2009-02-11
CA2322792C (fr) 2010-09-14
JP5931801B2 (ja) 2016-06-08
US6455283B1 (en) 2002-09-24
AU758353C (en) 1999-10-11
AU3075099A (en) 1999-10-11
EP1064382A2 (fr) 2001-01-03
US20080095773A1 (en) 2008-04-24
IL138488A (en) 2008-06-05
ES2313778T3 (es) 2009-03-01
US6620784B1 (en) 2003-09-16
JP4532733B2 (ja) 2010-08-25
DK2016951T3 (da) 2012-09-24
DE69939374D1 (de) 2008-10-02
US20030113870A1 (en) 2003-06-19
US20110319327A1 (en) 2011-12-29

Similar Documents

Publication Publication Date Title
EP1064382B1 (fr) Polypeptides homologues aux vegf et bmp1
EP1196186B1 (fr) Stimulation ou inhibition de l'angiogenese et de la cardiovascularisation avec des homologues de ligands et de recepteurs du facteur de necrose tumorale
JP2009019032A (ja) 血管形成及び心臓血管新生の促進又は阻害
WO1999014234A2 (fr) Stimulation ou inhibition de l'angiogenese et de la cardiovascularisation
AU5920099A (en) Promotion or inhibition of angiogenesis and cardiovascularization
AU2003204852B2 (en) Polypeptides homologous to VEGF and BMP1

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20001006

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

17Q First examination report despatched

Effective date: 20050204

17Q First examination report despatched

Effective date: 20050204

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 69939374

Country of ref document: DE

Date of ref document: 20081002

Kind code of ref document: P

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20080403035

Country of ref document: GR

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2313778

Country of ref document: ES

Kind code of ref document: T3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20090120

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20090525

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 18

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 19

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: LU

Payment date: 20180308

Year of fee payment: 20

Ref country code: CH

Payment date: 20180125

Year of fee payment: 20

Ref country code: NL

Payment date: 20180312

Year of fee payment: 20

Ref country code: GB

Payment date: 20180223

Year of fee payment: 20

Ref country code: FI

Payment date: 20180223

Year of fee payment: 20

Ref country code: DE

Payment date: 20180308

Year of fee payment: 20

Ref country code: DK

Payment date: 20180223

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AT

Payment date: 20180223

Year of fee payment: 20

Ref country code: IE

Payment date: 20180223

Year of fee payment: 20

Ref country code: MC

Payment date: 20180226

Year of fee payment: 20

Ref country code: SE

Payment date: 20180321

Year of fee payment: 20

Ref country code: IT

Payment date: 20180314

Year of fee payment: 20

Ref country code: FR

Payment date: 20180223

Year of fee payment: 20

Ref country code: GR

Payment date: 20180226

Year of fee payment: 20

Ref country code: BE

Payment date: 20180315

Year of fee payment: 20

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CY

Payment date: 20180216

Year of fee payment: 20

Ref country code: ES

Payment date: 20180403

Year of fee payment: 20

REG Reference to a national code

Ref country code: DE

Ref legal event code: R071

Ref document number: 69939374

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EUP

Effective date: 20190310

REG Reference to a national code

Ref country code: NL

Ref legal event code: MK

Effective date: 20190309

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: GB

Ref legal event code: PE20

Expiry date: 20190309

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK07

Ref document number: 405651

Country of ref document: AT

Kind code of ref document: T

Effective date: 20190310

REG Reference to a national code

Ref country code: IE

Ref legal event code: MK9A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20190309

Ref country code: IE

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20190310

REG Reference to a national code

Ref country code: BE

Ref legal event code: MK

Effective date: 20190310

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20201204

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF EXPIRATION OF PROTECTION

Effective date: 20190311